header logo image


Page 7«..6789..2030..»

Archive for the ‘Regenerative Medicine’ Category

CollPlant to trial 3D bioprinted breast implants in breakthrough animal study – 3D Printing Industry

Saturday, April 2nd, 2022

Regenerative medicine specialist CollPlant (NASDAQ: CLGN) has revealed that the 3D bioprinted breast tissues its developing are set to enter animal trials in the very near future.

Alongside the release of its FY 2021 results, which show that it generated $15.6 million in revenue, 155.7% more than the $6.1 million it reported in FY 2020, CollPlant announced that its bioprinted implants will enter in-vivo testing from Q2 2022.

Currently being developed with BICO firm CELLINK, the grafts are designed to gradually degrade and be replaced by native tissues, in a safer alternative to breast augmentation procedures. With further R&D, the companies believe their approach could be deployed in up to 2.2 million surgeries, enabling them to address a market worth an estimated $2.8 billion.

In the year ahead, we anticipate reaching development milestones for some of our leading programs, including the start of a large animal study for our 3D bioprinted regenerative breast implant program, said Yehiel Tal, CEO of CollPlant. We are also making continued progress with the development of bio-inks for 3D bioprinting applications, and with development of a photocurable dermal filler.

CollPlants FY 2021 financials

Although CollPlant hasnt provided a division-by-division breakdown of its annual revenue, its clear from what has been revealed, that its rate of growth is beginning to accelerate. In large part, the firms rapid rise in revenue last year was down to a $103 million bioprinting contract it signed with AbbVie company Allergan Aesthetics during March 2021.

As part of this agreement, the latter has begun to use CollPlants artificial collagen to create dermal and soft tissue fillers, and it could go on to develop two more products in future. The deal has also seen CollPlant receive an initial $14 million, and while this represented 90% of its FY 2021 revenue, it could gain another $89 million in milestone payments should any of Allergan Aesthetics products make it to market.

In addition to generating more revenue in FY 2021 than FY 2020, the company improved its profitability as well, raising its gross profit from $3.1 million to $13.6 million. This was primarily due to the fact that over this period, its cost of revenue fell from $3 million to $2 million, owing to expenses related to royalties, bio-ink and rhCollagen sales, and the ending of its deal with United Therapeutics in FY 2020.

Thanks largely to a registered direct offering in February 2021, which saw it raise $31.8 million in net proceeds and $6 million from the exercise of warrants, CollPlant was able to attract $38.8 million in financing across last year. As it happens, some $31.6 million of this funding was deployed immediately in short-term cash deposits, the likes of which generated up to $172,000 for the firm in FY 2020.

CollPlants breast implant trials

Since announcing the creation of its first regenerative breast implants in 2019, the firm has steadily sought to improve their clinical and commercial viability. The tissues themselves are made from patient fat cells and ECM components, as well as rhCollagen, CollPlants tobacco plant-grown alternative to animal or cadaver-sourced collagen.

In theory, once these grafts are injected into patients tissues, they foster the regeneration of native cells before slowly degrading, leaving behind no foreign contaminants. CollPlant says this procedure could offer a revolutionary alternative to silicone implants or fat transfer operations which come with a risk of adverse events, but they arent yet ready for market, and continue to undergo clinical trials.

One way the company has sought to accelerate the development of its bioprinted breast implants is via its recently-established partnership with CELLINK. When the collaboration was announced last month, CollPlant said that CELLINKs high-throughput bioprinters and expertise could enable it to overcome the hurdles facing its implants scalability.

That being said, the firm has also worked with 3D Systems before to develop breast reconstruction treatments for cancer survivors, and its possible that its current work will allow it to build on some of the findings it made as part of this project too.

In the case of its newly-announced trials, CollPlant has let little slip about their exact set up, but theyre understood to offer an opportunity to put its learnings from preclinical studies into practice, while taking a significant step forward in its breast implants R&D, in that their viability will now be tested in-vivo at scale.

Advances in breast implant bioprinting

Even though 3D bioprinting itself remains an emerging technology, a significant amount of progress has already been made in using it to produce viable breast implants. Earlier this year, Healshape raised $6.8 million towards the R&D of its patient-specific breast tissues, designed to treat those who have undergone a mastectomy.

In the past, Plcoskin has also announced plans to work with Yonsei University and LipoCoat with the aim of coming up with a novel 3D printed breast implant. In essence, the project was set up to combine LipoCoats lipid film coating technology and Plcoskins PCL-collagen coating approach, as a means of developing a uniquely-glazed graft that offers a reduced risk of infection or rejection.

Elsewhere, similar technologies are being developed to create all sorts of other tissues as well, ranging from the 3D bioprinted human tescticle cells produced at the University of British Columbia, to the bioprinted liver tissues of T&R Biofab.

To stay up to date with the latest 3D printing news, dont forget to subscribe to the 3D Printing Industry newsletter or follow us on Twitter or liking our page on Facebook.

For a deeper dive into additive manufacturing, you can now subscribe to our Youtube channel, featuring discussion, debriefs, and shots of 3D printing in-action.

Are you looking for a job in the additive manufacturing industry? Visit 3D Printing Jobs for a selection of roles in the industry.

Featured image shows a 3D bioprinted breast implant produced by CollPlant. Photo by Valerie Arad, CollPlant.

Go here to read the rest:
CollPlant to trial 3D bioprinted breast implants in breakthrough animal study - 3D Printing Industry

Read More...

Researchers Receive $2.5 Million Grant to Study Appendiceal Cancer – Newswise

Saturday, April 2nd, 2022

Newswise WINSTON-SALEM, N.C. March 31, 2022 Cancer of the appendix is rare, affecting only 1 in 100,000 people in the United States annually. However, because its so rare, theres limited research to help guide treatment decisions. But now, researchers at Atrium Health Wake Forest Baptists NCI-designated Comprehensive Cancer Center hope to change that with support from a $2.5 million grant from the National Cancer Institute.

Because the appendix is part of the gastrointestinal system, appendiceal cancers have traditionally been treated in the same way as colon cancer, said Lance Miller, associate professor of cancer biology at Wake Forest School of Medicine and co-principal investigator of the study. However, were learning that these cancers are molecularly very different. By increasing our molecular understanding of appendiceal cancer, we hope to have greater insight on how best to treat, which will lead to better outcomes.

According to Miller, appendiceal cancer is often diagnosed at late stages when it has already spread throughout the peritoneal cavity, the space within the abdomen that contains the stomach, liver and intestines. As a result, current treatment options are limited.

One treatment forpatients withappendiceal tumors with spread to the peritoneal cavityis cytoreductive surgery with hyperthermic intraperitoneal chemotherapy (CRS/HIPEC). TheCRS/HIPECprocedureinvolvessurgicallyremoving the cancerous tumors followed by the administration of heated chemotherapy directly into the abdomen to kill any residual cancer cells.

Wake Forest Baptist Medical Center was among the first hospitals in the U.S. to offer the procedure in 1991. The program is led by Dr. Edward Levine, professor of surgical oncology at Wake Forest School of Medicine and co-investigator of this study.

A major challenge is that some patients respond well to CRS/HIPEC, and some do not, said Dr. Konstantinos Votanopoulos, professor of surgery and director of the Wake Forest Organoid Research Center (WFORCE), a joint effort between the Wake Forest Baptist Comprehensive Cancer Center and the Wake Forest Institute for Regenerative Medicine (WFIRM) to tailor personalized therapy for patients. CRS/HIPEC is an aggressive, yet often effective, treatment in prolonging survival, but we dont know how patients will respond ahead of time.

Votanopoulos, who is also a co-principal investigator of the study, said the grant will support three objectives. One, researchers will build on previous research to develop a genetic test that will help identify patients who will benefit from CRS/HIPEC. Two, researchers will study gene expression patterns of high-grade tumors and how they impact survival. And three, the grant will support the use of patient-derived tumor organoids to study how mutations in the cancer might make a tumor more sensitive or resistant to certain chemotherapy drugs.

The creation of an organoid begins with a tissue biopsy of a tumor. Cells from this biopsy are then used to grow three-dimensional, patient-specific tumor organoids in the lab. By exposing the organoids to various chemotherapy drugs and observing their response, scientists can possibly predict how a patient will respond to treatment.

This research has the potential to create new possibilities for personalized medicine in the treatment of appendiceal cancer, Miller said.

Read more:
Researchers Receive $2.5 Million Grant to Study Appendiceal Cancer - Newswise

Read More...

Company Founded by Harvard Researchers to Launch Alternative Meat Product | News – Harvard Crimson

Saturday, April 2nd, 2022

Tender Foods, a food technology startup co-founded by four Harvard-affiliated researchers that produces alternative meats, is preparing for a product launch later this year.

The company, which produces plant-based meat spun from liquid polymers, is one of 27 startups launched in fiscal year 2021 to commercialize innovations from Harvard research labs. Tender Foods specializes in products that have a unique texture, structure, and ultimately taste, according to its founders.

A lot of the stuff that tries to mimic meat is textured, but its not fibrous, so its aligned and its a block of stuff, but its not individual fibers, said Luke A. MacQueen, one of the startups co-founders and a Harvard postdoctoral researcher in Bioengineering.

The Tender Foods products will better mimic the texture of real meat, MacQueen said.

MacQueen co-founded the company alongside three other Harvard affiliates: Bioengineering and Applied Physics professor Kevin K. Kit Parker, Grant M. Gonzalez 13, and SEAS researcher Christophe Chantre.

The fibers in Tender Foods meat are made using technology developed by Parker and his colleagues. The research group studied rotary jet-spinning, which uses centrifugal force to elongate liquid polymers into fibers. MacQueen likened the device to a cotton candy machine that works with different kinds of proteins.

The technology was initially used for various other purposes, including organ regeneration: in 2017, the researchers managed to spin nanofibers into biocompatible heart valves. Two years later, they showed the same could be done with gelatin scaffolds to hold animal muscle cells.

Every lesson learned from building tissues for regenerative medicine was applicable to building tissue to eat, Parker wrote in an email.

MacQueen said he is excited to see the variety of meats that might emerge from the startups technology.

When those fibers are spun and collected into a system, they can be tailored to be like the meat products people enjoy, whether they be as simple as a chicken breast or much more complicated layered structures, he said. Those can all be made in an artisanal way, starting with this very basic building block.

The research received funding from the Harvard Office of Technology Development and Harvards Wyss Institute for Biologically Inspired Engineering.

The first efforts to patent discoveries from my lab pertaining to meat were shot down by OTD around 2006, Parker wrote. We kept pushing.

MacQueen said he is excited to introduce Tender Foods products to the public.

As a young startup, weve had to kind of stay under the radar a little bit, but theres good things coming down the road, he said.

I ate some this morning, Parker added in an email. It was delicious.

Staff writer Felicia He can be reached at felicia.he@thecrimson.com.

Staff writer James R. Jolin can be reached at james.jolin@thecrimson.com.

Link:
Company Founded by Harvard Researchers to Launch Alternative Meat Product | News - Harvard Crimson

Read More...

FDA Grants Regenerative Medicine Advanced Therapy, Fast Track Designations to Novel CAR T-Cell Therapy for Relapsed, Refractory B-cell Non-Hodgkin…

Monday, January 17th, 2022

C-CAR039 showed positive efficacy and safety data in patients with relapsed or refractory B-cell non-Hodgkin lymphoma.

Officials with the FDA have granted both Regenerative Medicine Advanced Therapy (RMAT) Designation and Fast Track Designation to C-CAR039, a novel autologous bi-specific chimeric antigen receptor (CAR) T-cell therapy, for the treatment of patients with relapsed or refractory (r/r) diffuse large B cell lymphoma (DLBCL).

C-CAR039 targets both the CD19 and CD20 antigens, and early results from an investigator-led trial demonstrate positive efficacy and safety data in patients with r/r B-cell non-Hodgkin lymphoma. As of April 20, 2021, 34 patients received the therapy, 28 of whom were eligible for safety analyses and 27 of whom were evaluable for efficacy analyses. Patients median age was 55.5 years and 75% had cancer of Ann Arbor stage 3/4. Participants had a median of 3 prior lines of therapy and bridging therapy had been given to 17.9% of patients.

According to a press release, the best overall response rate was 92.6%, with a complete response rate of 85.2%. Patients had a median time to response of 1 month and at a median follow-up of 7 months, 74.1% of patients were still in complete remission. Furthermore, the 6-month estimated progression-free survival rate was 83.2%.

This is great news for CBMG that the FDA has granted C-CAR039 both RMAT and fast track designations based on its potential to increase objective and complete response rate in r/r DLBCL, said Tony Liu, chairman and CEO of Cellular Biomedicine Group, in the press release. The clinical data based on our clinical trials in China continue to support the hypothesis that C-CAR039 is the best-in-class CAR T asset for patients in this indication.

Cytokine release syndrome (CRS) was reported in 96% of patients, 92% of which was grade 1/2. Only 1 patient had grade 3 CRS. Immune effector cell-associated neurotoxicity syndrome occurred at grade 1 in 2 patients and no grade 2 or higher neurologic events were reported, according to the press release. The researchers will continue to evaluate patients with longer follow-up.

Separately, the FDA Office of Orphan Products Development granted an Orphan Drug Designation to C-CAR039 for the treatment of follicular lymphoma in June 2021. The Investigational New Drug application was cleared by the FDA on December 10, 2021.

We are working toward initiating 1b/2 trials for C-CAR039 in the US soon, Liu said in the press release. And we will work closely with the FDA to seek the best path forward to deliver the drug to patients in the US and EU.

REFERENCE

CBMG Receives FDA Regenerative Medicine Advanced Therapy and Fast Track Designations. News release. CBMG; January 12, 2022. Accessed January 13, 2022. https://www.cellbiomedgroup.com/newsroom/fda-rmat?lang=en

Read more here:
FDA Grants Regenerative Medicine Advanced Therapy, Fast Track Designations to Novel CAR T-Cell Therapy for Relapsed, Refractory B-cell Non-Hodgkin...

Read More...

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

Monday, January 17th, 2022

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 WHead of Business DevelopmentReports And Data | Web: http://www.reportsanddata.comDirect Line: +1-212-710-1370E-mail: [emailprotected]LinkedIn | Twitter | Blogs

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

SOURCE Reports And Data

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

Read More...

Europe Trailed US in Record Gene and Cell Therapy Funding in 2021 – Labiotech.eu

Monday, January 17th, 2022

Cell and gene therapy developers globally raised an all-time annual record in 2021. However, European firms missed out on the funding growth.

Companies around the world developing cell and gene therapies raised 20.1B ($23.1B) over 2021, said the advanced therapy advocacy organization the Alliance for Regenerative Medicine (ARM) in a briefing this week. This bumper catch beat 2020s total of 17.3B ($19.9B) by 16%.

The growth from 2020 to 2021 was primarily driven by companies in the US. With a fresh 15.7B ($18B) in the bank, US-based companies saw an impressive 53% jump in investments compared to 2020. In contrast, their European counterparts raised 2.9B ($3.3B), 8% less funding than in 2020.

Both European and US gene and cell therapy players had seen record funding growth in 2020 compared to 2019, said Stephen Majors, ARMs Director of Public Affairs. However, its too early to establish why European and Asian companies havent matched the rapid cash growth seen in the US over 2021.

Its something well watch closely over the next year to determine what the causes may be and whether they are region-specific, said Majors.

Nonetheless, the funding numbers need to be interpreted in the correct context, said Antoine Papiernik, Chairman and Managing Partner of the venture capital (VC) firm Sofinnova Partners. European contributions to the field of cell and gene therapy remain immense.

Its not about how much you raise in one year; its about the level of expertise, competencies, and technologies, said Papiernik. These are the fundamentals for long-term excellence and growth, which we strongly believe in.

If there is one area where Europe is, without a doubt, on par with the US, its in new modalities, which include gene and cell therapies.

Of the various funding sources going to cell and gene therapies, VC funding increased the most in 2021, with a huge 73% jump to 8.5B ($9.8B). This trend mirrored the deluge in life sciences VC funding in the last year.

Simultaneously, gene and cell therapy companies were hit by struggling stock markets affecting the rest of the biotech sector. This mismatch is creating a bulge in funding for VC firms and potentially limiting exit options.

Inflation concerns made it particularly difficult for smaller, early-stage companies that are not yet profitable, said Majors. If inflation concerns subside in 2022, and with positive data readouts, we could see stronger performance for biotech public equities.

When the total is broken down by the types of technology getting funded, cell therapies in immuno-oncology such as CAR-T cell therapies saw the biggest funding increase: a jump of 26% since 2020. This was followed by gene therapy firms with 14% more incoming cash, and tissue engineering players, whose investments went up by 10%.

Cell therapy companies outside of immuno-oncology experienced a tighter year for financing in 2021 than in 2020, taking in 15% less funding at 1.7B ($2B). However, Majors told me that funding in this field has regularly fluctuated in the last several years.

The decrease over 2021 is not an outlier in comparison to historical trends, Majors noted. Due to the smaller size of this technology segment, just one or two financing deals can have a large impact on total financing on an annual basis.

Another important trend in the ARMs report was the rising importance of gene-editing technology. Of the total gene therapy financing, 45% was raised by companies developing gene-editing technology, up from 38% in 2018.

Investor interest in gene editing has been buoyed by clinical successes from frontrunner gene therapy players in the last year. One example from June 2021 was the promising performance of an in vivo CRISPR treatment developed by Intellia Therapeutics and Regeneron in patients with the rare disease transthyretin amyloidosis.

Gene-editing firms CRISPR Therapeutics and Vertex Pharmaceuticals are causing excitement with progress in tackling the blood disorder sickle cell disease. They are gunning to file for approval of their CRISPR gene-edited therapy for this condition in late 2022.

Investors have taken note of these early successes and see this approachs potential to treat a wide range of diseases, said Majors. Also, as this technology continues to progress, the number of companies with at least one clinical or preclinical asset in gene editing continues to rise.

Another outcome to look forward to for gene and cell therapy in 2022 is a potential record number of drug approvals. A bunch of gene therapy hopefuls including GenSight, uniQure, and BioMarin are poised to bring their candidates to the regulatory finish line in the US and Europe.

The EMA is slated to make decisions on therapies targeting aromatic l-amino acid decarboxylase deficiency, Leber hereditary optic neuropathy, and two types of hemophilia, said Majors. By the end of 2022, the number of EMA-approved gene therapies for rare diseases may have doubled from a year earlier.

However, some of the major hurdles for the field will likely be the delivery of gene and cell therapies to their target in the body as well as deciding the right dosage. The manufacture of these complex therapies is also a big bottleneck that many startups aim to tackle.

Additionally, the withdrawal of bluebird bios gene therapy from Germany in May 2021 over pricing disagreements demonstrates that regulatory approval is just the beginning for developers of gene and cell therapies. Their pricing strategy will need to walk the tightrope of making a profit while avoiding clashes with healthcare systems.

In any case, European companies will continue to play a strong role in the evolution of the cell and gene therapy sphere.

Lets not forget that the first gene therapy to be brought to the market was European, said Papiernik, referring to the gene therapy Strimvelis, which was sold by GlaxoSmithKline to Orchard Therapeutics in 2018.

Europe continues to excel in the development of gene and cell therapies and never has there been more opportunities for investment.

Cover image via Elena Resko. Inline images via the Alliance for Regenerative Medicine

Read this article:
Europe Trailed US in Record Gene and Cell Therapy Funding in 2021 - Labiotech.eu

Read More...

NIH researchers develop first stem cell model of albinism to study related eye conditions – National Institutes of Health

Monday, January 17th, 2022

News Release

Tuesday, January 11, 2022

Use of patient-derived stem cells will enable high-throughput drug screening for potential therapeutics.

Researchers at the National Eye Institute (NEI) have developed the first patient-derived stem cell model for studying eye conditions related to oculocutaneous albinism (OCA). The models development is described in the January issue of the journal Stem Cell Reports. NEI is part of the National Institutes of Health.

This disease-in-a-dish system will help us understand how the absence of pigment in albinism leads to abnormal development of the retina, optic nerve fibers, and other eye structures crucial for central vision, said Aman George, Ph.D., a staff scientist in the NEI Ophthalmic Genetics and Visual Function Branch, and the lead author of the report.

OCA is a set of genetic conditions that affects pigmentation in the eye, skin, and hair due to mutation in the genes crucial to melanin pigment production. In the eye, pigment is present in the retinal pigment epithelium (RPE), and aids vision by preventing the scattering of light. The RPE is located right next to the eyes light-sensing photoreceptors and provides them nourishment and support. People with OCA lack pigmented RPE and have an underdeveloped fovea, an area within the retina that is crucial for central vision. The optic nerve carries visual signals to the brain.

People with OCA have misrouted optic nerve fibers. Scientists think that RPE plays a role in forming these structures and want to understand how lack of pigment affects their development.

Animals used to study albinism are less than ideal because they lack foveae, said Brian P. Brooks, M.D., Ph.D., NEI clinical director and chief of the Ophthalmic Genetics and Visual Function Branch. A human stem cell model that mimics the disease is an important step forward in understanding albinism and testing potential therapies to treat it.

To make the model, researchers reprogrammed skin cells from individuals without OCA and people with the two most common types of OCA (OCA1A and OCA2) into pluripotent stem cells (iPSCs). The iPSCs were then differentiated to RPE cells. The RPE cells from OCA patients were identical to RPE cells from unaffected individuals but displayed significantly reduced pigmentation.

The researchers will use the model to study how lack of pigmentation affects RPE physiology and function. In theory, if fovea development is dependent on RPE pigmentation, and pigmentation can be somehow improved, vision defects associated with abnormal fovea development could be at least partially resolved, according to Brooks.

Treating albinism at a very young age, perhaps even prenatally, when the eyes structures are forming, would have the greatest chance of rescuing vision, said Brooks. In adults, benefits might be limited to improvements in photosensitivity, for example, but children may see more dramatic effects.

The team is now exploring how to use their model for high-throughput screening of potential OCA therapies.

NEI leads the federal governments research on the visual system and eye diseases. NEI supports basic and clinical science programs to develop sight-saving treatments and address special needs of people with vision loss. For more information, visit https://www.nei.nih.gov.

About the National Institutes of Health (NIH):NIH, the nation's medical research agency, includes 27 Institutes and Centers and is a component of the U.S. Department of Health and Human Services. NIH is the primary federal agency conducting and supporting basic, clinical, and translational medical research, and is investigating the causes, treatments, and cures for both common and rare diseases. For more information about NIH and its programs, visit http://www.nih.gov.

NIHTurning Discovery Into Health

Aman George, Ruchi Sharma, Tyler Pfister, Mones Abu-Asab, Nathan Hotaling, Devika Bose, Charles DeYoung, Justin Chang, David R. Adams, Tiziana Cogliati, Kapil Bharti, Brian P. Brooks. In Vitro Disease Modeling of Oculocutaneous Albinism Type I and II Using Human Induced Pluripotent Stem Cell-Derived Retinal Pigment Epithelium (2022). doi: 10.1016/j.stemcr.2021.11.01.https://www.cell.com/stem-cell-reports/fulltext/S2213-6711(21)00597-X.

###

Read the original post:
NIH researchers develop first stem cell model of albinism to study related eye conditions - National Institutes of Health

Read More...

Scientists uncover new information about cellular death process, previously thought to be irreversible | UIC Today – UIC Today

Monday, January 17th, 2022

A study published by researchers at the University of Illinois Chicago describes a new method for analyzing pyroptosis the process of cell death that is usually caused by infections and results in excess inflammation in the body and shows that process, long thought to be irreversible once initiated, can in fact be halted and controlled.

The discovery, which is reported in Nature Communications, means that scientists have a new way to study diseases that are related to malfunctioning cell death processes, like some cancers, and infections that can be complicated by out-of-control inflammation caused by the process. These infections include sepsis, for example, and acute respiratory distress syndrome, which is among the major complications of COVID-19 illness.

Pyroptosis is a series of biochemical reactions that uses gasdermin, a protein, to open large pores in the cell membrane and destabilize the cell. To understand more about this process, the UIC researchers designed an optogenetic gasdermin by genetically engineering the protein to respond to light.

The cell death process plays an important role in the body, in both healthy states and unhealthy ones, but studying pyroptosis which is a major type of cell death has been challenging, said Gary Mo, UIC assistant professor in the department of pharmacology and regenerative medicine and the department of biomedical engineering at the College of Medicine.

Mo said that methods to examine the pyroptosis mechanisms at play in live cells are difficult to control because they are initiated by unpredictable pathogens, which in turn have disparate effects in different cells and people.

Our optogeneticgasderminallowed us to skipovertheunpredictablepathogen behavior and the variable cellular responsebecause itmimics at the molecular level what happens in the celloncepyroptosisis initiated,Mo said.

The researchersappliedthis tool andusedflorescent imaging technologyto precisely activategasderminincell experimentsand observethe pores under various circumstances. They discovered that certainconditions, like specificconcentrationsof calcium ions, for example, triggered the pores to close within only tens of seconds.

This automatic response toexternalcircumstances provides evidence thatpyroptosisdynamically self-regulates.

This showed us that this form of cell death is not a one-way ticket. The process isactually programmedwith acancel button, anoff-switch, Mo said. Understanding how to control this process unlocks new avenues for drug discovery, and now we can find drugs that work for both sides it allows us to think about tuning, either boosting or limiting, this type of cell death in diseases, where we could previously only remove this important process.

Co-authors of theNature Communicationspaper,GasderminD Pores Are Dynamically Regulated by Local Phosphoinositide Circuitry,areAna Santa Cruz Garcia, KevinSchnurand Asrar Malik,all ofUIC.

The research was funded with grants from the National Institutes of Health (P01HL060678, R01HL090152, R01HL152515, T32HL007820, P01HL151327).

More:
Scientists uncover new information about cellular death process, previously thought to be irreversible | UIC Today - UIC Today

Read More...

Founders’ Day Speaker: Breakthrough Discoveries Lead to Real World Applications – Illinois Wesleyan University

Monday, January 17th, 2022

Jan. 13, 2022

Health and innovative thinking was the focus of this years Founders Day Convocation at Illinois Wesleyan University.

A recording of the virtual Convocation may be viewed here.

Illinois Wesleyan President S. Georgia Nugent spoke of the motives that led to the founding of the University in 1850 and their relevance today, as well as the annual intellectual theme of Health, Healing and Humanity.

A theme that obviously could not be more relevant today, as we see these three inextricably intertwined in the context of the Coronavirus pandemic, she said.

Biomedical researcher William Murphy, a 1998 IWU Physics and math graduate, gave the keynote address titled Mimicking Nature to Create New Technology.

Murphy spoke about how his experience as a Titan helped shape his future career in biotechnology. He shared examples of his research efforts to create regenerative medicine based on materials already found in nature.

You are fortunate to be enrolled at a yes, and institution, said Murphy. You dont have to choose to become only a physicist, or only a chemist, or only a business student. You can also be a baseball player or a musician. One can engage in all of these opportunities at once to build what will become the foundation for your lifes journey.

He reminded students that every course of study can make an impact on the world.

I hope Ive convinced you today that there is so much more to discover and leverage in nature and that all disciplines can contribute to the future of biotechnology, he said. Your IWU education is preparing you wonderfully to make breakthrough discoveries and turn them into real-world products.

By Julia Perez

Read this article:
Founders' Day Speaker: Breakthrough Discoveries Lead to Real World Applications - Illinois Wesleyan University

Read More...

Coronavirus FAQ: Why are some folks hacking home COVID tests by swabbing their throat? – Wisconsin Public Radio News

Monday, January 17th, 2022

Lots of folks on Twitter say that swabbing your throat in addition to your nose may be a better way of using your COVID-19 testing kit to detect the omicron variant. Is this true?

If you've used a do-it-yourself COVID-19 home test in the U.S. the "antigen" rapid tests that promise results in 15 minutes or so you know the drill. You typically swirl a cotton swab around in your nostrils, mix it with some liquid and then drop it on a test strip to await the results: positive or negative for the coronavirus.

But in recent weeks, there has been an online debate about where to stick that cotton swab. Although the directions specify a nasal swab on U.S. products, some medical professionals believe the test is more effective at detecting the coronavirus, and specifically the omicron variant, if the kit's swab is used in the throat and/or cheek in addition to the nose.

Why did this hack emerge and is there any medical science to back it up?

The idea of a throat swab is not in and of itself a radical step. Even though antigen test kits in the U.S. are designed for a nasal swab, home tests are designed for a throat swab in other countries in Canada and the U.K., for example.

The idea of improvising with a throat swab is connected to the omicron variant, which was identified in late November and has swept the world. Researchers increasingly believe omicron may replicate in the throat before the nose.

That was one of the findings of a study conducted at the University of Cape Town in South Africa. Researchers examined the ability of PCR tests to detect the variant by comparing diagnostic tests for 382 symptomatic COVID-19 patients who weren't hospitalized. In patients with the delta variant of the coronavirus, saliva swabs detected the virus 71% of the time, while nasal swabs found it 100% of the time. But in patients with omicron, researchers found the reverse: Saliva swabs detected the virus 100% of the time, while nasal swabs caught it 86% of the time.

The research has not been peer reviewed. And the tests used in the study were PCR tests, not antigen tests. (PCR tests are the gold standard and are administered typically in a health care facility; antigen tests are less precise and can be done at home. See this story for details on the differences.)

Nonetheless, the South African study has led some epidemiologists and immunologists in the U.S. to experiment with antigen tests by swabbing their throats or cheeks in addition to the nose when administering a self-test.

That's not an authorized use of the kits, reminds the Food and Drug Administration, which regulates the kits: "FACT: When it comes to at-home rapid antigen #COVID19 tests, those swabs are for your nose and not your throat," it said in a tweet issued on Jan. 7.

And even proponents of the throat swab stop short of endorsing the practice for the public without a green light from the FDA.

One of the most often quoted voices in favor of throat swabbing is Dr. Michael Mina, formerly an epidemiologist and immunologist at the Harvard T.H. Chan School of Public Health and now the chief science officer at the testing company eMed. He has tweeted: "We should be rigorously looking into throat swabs to add some level of sensitivity" and "Throat swab + nasal may improve chances a swab picks up virus."

Stay informed with WPR's email newsletter.

But Mina has gone on to tweet that the FDA was right to warn the public not to go against manufacturer's directions: "Telling US public not to go against directions is the *right* thing to do."

"The tests haven't been designed to do [throat swabs], so we don't know whether there are false positives or negatives," agrees Dr. Jill Weatherhead, assistant professor of adult and pediatric infectious diseases at Baylor College of Medicine. "It logically makes sense that you'd want to do it, but that's not how [the tests have] been developed or tested."

One potential problem could be the thicker viscosity of throat saliva, says Dr. Yuka Manabe, associate director of global health research and innovation and a professor of medicine at the Johns Hopkins Bloomberg School of Public Health. It's possible the sample would need to be filtered first, she says, to produce an accurate result.

That's not necessarily a critical concern, says Michael Daignault, an emergency physician who serves as chief medical adviser to Reliant Health Services, a medical testing company. He says that the nature of throat saliva could simply cause the test to display a message that the result is invalid rather than a false positive.

Another obvious potential problem, Manabe points out, is the length of the swab in some of the kits: "The swab that you use doesn't have a very long stick because it's just meant to be used in the front of your nose. So for you to get that into the back of your throat would not be very easy, truthfully, for some of the kits, depending on the length of the swab."

The public debate about the best way to swab has prompted a growing chorus of voices to call for new studies and a revamping of the kits and/or the instructions.

That kind of change is what Michal Tal is hoping for. Tal, an instructor at Stanford University's Institute for Stem Cell Biology and Regenerative Medicine and a visiting scientist at the Massachusetts Institute of Technology, is a proponent of throat swabs. Before seeing anyone in person, she asks that they take a coronavirus test and swab not only their nose but their cheeks, the roof of their mouth, under their tongue and if they don't gag easily their throat near the tonsils.

"I'm feeling very frustrated that the FDA and CDC didn't jump on this and try to make more rapid changes," says Tal. "The virus is always two steps ahead and we don't adapt."

At a congressional hearing Monday, the acting head of the FDA, Dr. Janet Woodcock, noted that the National Institutes of Health has helped accelerate the authorization of new home tests so that they can get the FDA's signoff in just one or two days. But she said it may take a while for companies to change their test configurations to include larger swabs for the throat.

In the meantime, she warned people not to swab their throats with the current devices, which are designed as nasal swabs. "They may stab themselves," she said. "That would not be good."

Sheila Mulrooney Eldred is a freelance health journalist in Minneapolis. She has written about COVID-19 for many publications, including The New York Times, Kaiser Health News, Medscape and The Washington Post. More at sheilaeldred.pressfolios.com. On Twitter: @milepostmedia.

The rest is here:
Coronavirus FAQ: Why are some folks hacking home COVID tests by swabbing their throat? - Wisconsin Public Radio News

Read More...

Global Sports Medicine & Physiotherapy Market Research Report (2021 to 2027) – by Product, Therapy Equipment, Application, End-user and Region -…

Monday, January 17th, 2022

DUBLIN, Jan. 14, 2022 /PRNewswire/ -- The "Sports Medicine & Physiotherapy Market Research Report by Product, by Therapy Equipment, by Application, by End User, by Region - Global Forecast to 2027 - Cumulative Impact of COVID-19" report has been added to ResearchAndMarkets.com's offering.

The Global Sports Medicine & Physiotherapy Market size was estimated at USD 8,245.72 million in 2020, is expected to reach USD 8,944.13 million in 2021, and is projected to grow at a CAGR of 8.83% to reach USD 14,915.43 million by 2027.

Market Statistics:

The report provides market sizing and forecast across five major currencies - USD, EUR GBP, JPY, and AUD. It helps organization leaders make better decisions when currency exchange data is readily available. In this report, the years 2018 and 2019 are considered historical years, 2020 as the base year, 2021 as the estimated year, and years from 2022 to 2027 are considered the forecast period.

Competitive Strategic Window:

The Competitive Strategic Window analyses the competitive landscape in terms of markets, applications, and geographies to help the vendor define an alignment or fit between their capabilities and opportunities for future growth prospects. It describes the optimal or favorable fit for the vendors to adopt successive merger and acquisition strategies, geography expansion, research & development, and new product introduction strategies to execute further business expansion and growth during a forecast period.

FPNV Positioning Matrix:

The FPNV Positioning Matrix evaluates and categorizes the vendors in the Sports Medicine & Physiotherapy Market based on Business Strategy (Business Growth, Industry Coverage, Financial Viability, and Channel Support) and Product Satisfaction (Value for Money, Ease of Use, Product Features, and Customer Support) that aids businesses in better decision making and understanding the competitive landscape.

Market Share Analysis:

The Market Share Analysis offers the analysis of vendors considering their contribution to the overall market. It provides the idea of its revenue generation into the overall market compared to other vendors in the space. It provides insights into how vendors are performing in terms of revenue generation and customer base compared to others. Knowing market share offers an idea of the size and competitiveness of the vendors for the base year. It reveals the market characteristics in terms of accumulation, fragmentation, dominance, and amalgamation traits.

Company Usability Profiles:

The report profoundly explores the recent significant developments by the leading vendors and innovation profiles in the Global Sports Medicine & Physiotherapy Market, including Acumed LLC, Arthrex, Inc., Bauerfeind AG, Biotek, Breg Inc, Carestream Health, Conmed Corporation, Cramer Products, DePuy Synthes, DJO Global, Inc., Include Health Inc., iTrace Biomedical Inc., KARL STORZ, KATOR LLC, MedShape, Medtronic, MTF Biologics, Mueller Sports, Inc., Performance Health International Limited, PolyPhysics, Inc., Quadrant Biosciences, RTI Surgical, RoG Sports Medicine, Smith & Nephew PLC, Stryker Corporation, Wright Medical Group N.V., Wright Medical Technology, Zimmer Biomet, and Ossur.

The report provides insights on the following pointers:1. Market Penetration: Provides comprehensive information on the market offered by the key players2. Market Development: Provides in-depth information about lucrative emerging markets and analyze penetration across mature segments of the markets3. Market Diversification: Provides detailed information about new product launches, untapped geographies, recent developments, and investments4. Competitive Assessment & Intelligence: Provides an exhaustive assessment of market shares, strategies, products, certification, regulatory approvals, patent landscape, and manufacturing capabilities of the leading players5. Product Development & Innovation: Provides intelligent insights on future technologies, R&D activities, and breakthrough product developments

The report answers questions such as:1. What is the market size and forecast of the Global Sports Medicine & Physiotherapy Market?2. What are the inhibiting factors and impact of COVID-19 shaping the Global Sports Medicine & Physiotherapy Market during the forecast period?3. Which are the products/segments/applications/areas to invest in over the forecast period in the Global Sports Medicine & Physiotherapy Market?4. What is the competitive strategic window for opportunities in the Global Sports Medicine & Physiotherapy Market?5. What are the technology trends and regulatory frameworks in the Global Sports Medicine & Physiotherapy Market?6. What is the market share of the leading vendors in the Global Sports Medicine & Physiotherapy Market?7. What modes and strategic moves are considered suitable for entering the Global Sports Medicine & Physiotherapy Market?

Key Topics Covered:

1. Preface

2. Research Methodology

3. Executive Summary

4. Market Overview

5. Market Insights5.1. Market Dynamics5.1.1. Drivers5.1.1.1. Rising incidence of sports injuries worldwide5.1.1.2. Growing demand for minimally invasive surgeries5.1.1.3. Rapid introduction of new products toward treatment modalities5.1.2. Restraints5.1.2.1. Relatively high cost of implants and other devices5.1.3. Opportunities5.1.3.1. Ongoing developments in regenerative medicine5.1.3.2. Surge in adoption of computer assisted robotic surgeries to reduce recovery time5.1.4. Challenges5.1.4.1. Lack of skilled expertise in the field5.2. Cumulative Impact of COVID-19

6. Sports Medicine & Physiotherapy Market, by Product6.1. Introduction6.2. Accessories6.3. Body Reconstruction Products6.3.1. Arthroscopy Devices6.3.2. Fracture and Ligament Repair Products6.3.3. Implants6.3.4. Orthobiologics6.3.5. Prosthetic Devices6.4. Body Support and Recovery Products6.4.1. Braces and Supports6.4.2. Compression Clothing6.4.3. Physiotherapy Equipment

7. Sports Medicine & Physiotherapy Market, by Therapy Equipment7.1. Introduction7.2. Accessories7.2.1. Physiotherapy Furniture7.2.2. Physiotherapy Tapes & Bandages7.3. Equipment7.3.1. Combination Therapy Equipment7.3.2. Continuous Passive Motion Therapy Equipment7.3.3. Cryotherapy Equipment7.3.4. Electrotherapy Equipment7.3.5. Exercise Therapy Equipment7.3.6. Laser Therapy Equipment7.3.7. Magnetic & Pressure Therapy Equipment7.3.8. Shockwave Therapy Equipment7.3.9. Traction Therapy Equipment7.3.10. Ultrasound Equipment

8. Sports Medicine & Physiotherapy Market, by Application8.1. Introduction8.2. Back & Spine Injuries8.3. Cardiovascular & Pulmonary8.4. Elbow & Wrist Injuries8.5. Foot & Ankle Injuries8.6. Gynecological8.7. Hip & Groin Injuries8.8. Knee Injuries8.9. Musculoskeletal8.10. Neurological8.11. Pediatric8.12. Shoulder Injuries

9. Sports Medicine & Physiotherapy Market, by End User9.1. Introduction9.2. Ambulatory Surgery Centres9.3. Home Care Settings9.4. Hospitals9.5. Physiotherapy Centres & Clinics

10. Americas Sports Medicine & Physiotherapy Market10.1. Introduction10.2. Argentina10.3. Brazil10.4. Canada10.5. Mexico10.6. United States

11. Asia-Pacific Sports Medicine & Physiotherapy Market11.1. Introduction11.2. Australia11.3. China11.4. India11.5. Indonesia11.6. Japan11.7. Malaysia11.8. Philippines11.9. Singapore11.10. South Korea11.11. Taiwan11.12. Thailand

12. Europe, Middle East & Africa Sports Medicine & Physiotherapy Market12.1. Introduction12.2. France12.3. Germany12.4. Italy12.5. Netherlands12.6. Qatar12.7. Russia12.8. Saudi Arabia12.9. South Africa12.10. Spain12.11. United Arab Emirates12.12. United Kingdom

13. Competitive Landscape13.1. FPNV Positioning Matrix13.1.1. Quadrants13.1.2. Business Strategy13.1.3. Product Satisfaction13.2. Market Ranking Analysis13.3. Market Share Analysis, By Key Player13.4. Competitive Scenario13.4.1. Merger & Acquisition13.4.2. Agreement, Collaboration, & Partnership13.4.3. New Product Launch & Enhancement13.4.4. Investment & Funding13.4.5. Award, Recognition, & Expansion

14. Company Usability Profiles14.1. Acumed LLC14.2. Arthrex, Inc.14.3. Bauerfeind AG14.4. Biotek14.5. Breg Inc14.6. Carestream Health14.7. Conmed Corporation14.8. Cramer Products14.9. DePuy Synthes14.10. DJO Global, Inc.14.11. Include Health Inc.14.12. iTrace Biomedical Inc.14.13. KARL STORZ14.14. KATOR LLC14.15. MedShape14.16. Medtronic14.17. MTF Biologics14.18. Mueller Sports, Inc.14.19. Performance Health International Limited14.20. PolyPhysics, Inc.14.21. Quadrant Biosciences14.22. RTI Surgical14.23. RoG Sports Medicine14.24. Smith & Nephew PLC14.25. Stryker Corporation14.26. Wright Medical Group N.V.14.27. Wright Medical Technology14.28. Zimmer Biomet14.29. Ossur

15. Appendix

For more information about this report visit https://www.researchandmarkets.com/r/d6ieqy

Media Contact:

Research and Markets Laura Wood, Senior Manager [emailprotected]

For E.S.T Office Hours Call +1-917-300-0470 For U.S./CAN Toll Free Call +1-800-526-8630 For GMT Office Hours Call +353-1-416-8900

U.S. Fax: 646-607-1907 Fax (outside U.S.): +353-1-481-1716

SOURCE Research and Markets

Follow this link:
Global Sports Medicine & Physiotherapy Market Research Report (2021 to 2027) - by Product, Therapy Equipment, Application, End-user and Region -...

Read More...

Evolved by Nature Announces IDE Approval from the FDA to Initiate a Pivotal Clinical Trial for its first Activated Silk Dermal Filler – PRNewswire

Monday, January 17th, 2022

BOSTON, Jan. 12, 2022 /PRNewswire/ --Biotechnology company Evolved by Nature announced today that sister company Silk Medical Aesthetics has received Investigational Device Exemption (IDE) approval from the FDA to initiate a pivotal clinical trial for its new dermal filler for the correction of moderate to severe facial wrinkles and folds, such as nasolabial folds.

The new filler leverages Evolved by Nature's all-natural Activated Silk protein technology platform, infusing it within a crosslinked hyaluronic acid gel with lidocaine. It isdesigned to be biodegradable and reversible for use in soft tissue augmentation such as for wrinkle filling and volumizing. The filler is the first in a differentiated product line being developed to provide both the patient and physician with new options in aesthetic medicine to address needs from facial skin smoothing and contouring to enhancing skin attributes for improved patient outcomes.

The randomized, controlled, blinded, prospective pivotal clinical trial will establish the Activated Silk dermal filler's safety and efficacy versus a commercially FDA-approved available control. Silk Medical Aesthetics will initiate the study with multiple centers in Q1 2022, and trial results will be submitted in a future Premarket Approval (PMA) application to the FDA to support regulatory approval.

Primary endpoint data for the feasibility clinical trial on the Activated Silk filler, announced in late 2021, demonstrated a favorable safety profile with high patient satisfaction and clinically meaningful improvement in NLF severity. Zero device-related adverse events were reported, and 83.7% of patients had a clinically meaningful improvement in wrinkle severity at the 13-week primary timepoint. 100% of clinicians and patients reported improvement on the Global Aesthetic Improvement Scale (GAIS).

Evolved by Nature CEO, Dr. Greg Altman, commented, "We've always known that Activated Silk protein technology has the potential to dramatically restore or improvebarrier function, on everything from skin to textiles used by the fashion industry, and more. This IDE approval is a tremendous benchmark as we rapidly expand our development pipeline, exploring and developing medical devices, therapeutics, and bioactive molecules for regenerative medicine."

About Evolved by Nature: Founded in 2013 in Boston, MA, Evolved by Nature is a biotechnology company that creates renewably sourced solutions to human health needs. Activated Silk biotechnology leverages natural silk protein to create sustainable molecules that protect, repair, and enhance the barrier function of anything with a surface. Enabling the next generation of products that advance the health of people and the planet, Evolved by Nature has unlocked breakthrough applications for Activated Silk biotechnology within textiles, personal care, aesthetic and medical treatments, therapeutics and more, with limitless possibilities. http://www.evolvedbynature.com

About Silk Medical Aesthetics: Silk Medical Aesthetics is a Boston-based company on a mission to create the next-generation dermal filler platform by leveraging the power of natural silk. A sister company of biotechnology company Evolved by Nature, Silk Medical Aesthetics was founded by silk experts Drs. Greg Altman and Rebecca Lacouture in 2018, received more than $18 million in Series A and B financing, and is backed by a diverse group of investors. http://www.silkmedicalaesthetics.com

SOURCE Evolved By Nature

See the original post here:
Evolved by Nature Announces IDE Approval from the FDA to Initiate a Pivotal Clinical Trial for its first Activated Silk Dermal Filler - PRNewswire

Read More...

TrialSpark licenses sprifermin, an investigational first-in-class disease modifying treatment for osteoarthritis, from Merck KGaA, Darmstadt, Germany…

Monday, January 17th, 2022

Sprifermin promotes cartilage growth, and could be a potential disease modifying treatment for osteoarthritis.

"We believe sprifermin has the potential to be the first disease modifying therapy approved for the millions of patients suffering from OA," said Gavin Corcoran, Chief Medical Officer at TrialSpark. "Despite recent advances in our understanding of the pathogenesis of OA, clinical development has remained behind other rheumatic diseases. TrialSpark's proprietary clinical trial engine is uniquely positioned to address historical challenges in OA development including long studies requiring many patients, designing optimal pain endpoints, and identifying key patient subgroups most likely to benefit."

TrialSpark's ability to leverage its tech-enabled trial engine to run faster and more efficient drug development enables it to pursue programs in indications - such as OA - that have historically required the longest, most expensive clinical trials. High Line Bio will leverage TrialSpark's proprietary tech-enabled clinical trial engine to develop sprifermin in OA using a data-driven approach to identifying novel biomarkers, endpoints, and patient subgroups most likely to benefit from the therapy. TrialSpark is also partnering with industry leaders such as SomaLogic to leverage precision genomics and proteomics platforms to identify key biomarkers and stratify patients using synovial fluid samples from prior clinical studies using AI and machine-learning approaches. Beyond sprifermin, High Line Bio also plans in-license additional complementary products to build a differentiated pipeline focused on OA and regenerative medicine.

This transaction is in line with TrialSpark's strategy of creating new companies across key therapeutic areas like inflammatory and autoimmune disease. To date, TrialSpark has successfully in-licensed multiple assets, deploying capital and supporting programs across a broad range of therapeutic areas including CNS, dermatology, and inflammation as part of its mission to bring new treatments to patients faster and more efficiently.

As part of its business development and investments strategy, TrialSpark is using balance sheet capital to acquire or license assets, leveraging its in-house clinical development engine to run trials significantly faster and more efficiently than industry. In addition to conventional asset licensing and acquisition, TrialSpark transactions can also include equity investments in both private and public biopharma companies, strategic collaborations to jointly fund and develop assets, and alternative structures including syndication with other investors.

TrialSpark is actively pursuing partnerships with both small biotech and larger pharma companies through flexible and creative collaboration structures, maximizing the potential value of drug candidates for all stakeholders through faster and more efficient clinical development programs.TrialSpark is focused on early to mid-stage clinical assets across a range of therapeutic areas, with a focus on chronic diseases that are treated outside of a hospital setting.

About Merck KGaA, Darmstadt, Germany Merck KGaA, Darmstadt, Germany, a leading science and technology company, operates across healthcare, life science and electronics. Around 58,000 employees work to make a positive difference to millions of people's lives every day by creating more joyful and sustainable ways to live. From advancing gene editing technologies and discovering unique ways to treat the most challenging diseases to enabling the intelligence of devices the company is everywhere. In 2020, Merck KGaA, Darmstadt, Germany, generated sales of 17.5 billion in 66 countries.

The company holds the global rights to the name and trademark "Merck" internationally. The only exceptions are the United States and Canada, where the business sectors of Merck KGaA, Darmstadt, Germany operate as EMD Serono in healthcare, MilliporeSigma in life science, and EMD Performance Materials. Since its founding 1668, scientific exploration and responsible entrepreneurship have been key to the company's technological and scientific advances. To this day, the founding family remains the majority owner of the publicly listed company.

About TrialSpark TrialSpark is a technology-driven pharma company that runs end-to-end clinical trials, focused on bringing new treatments to patients faster and more efficiently. TrialSpark has built a technology platform that optimizes all aspects of a clinical trial, enabling more efficient trial design, faster trial completion, and higher trial data quality. TrialSpark in-licenses and co-develops drug programs through in-house development, joint ventures, and NewCos. TrialSpark is backed by leading investors such as Sam Altman, Lachy Groom, Michael Moritz, Casdin Capital, Sequoia Capital, Thrive Capital, Dragoneer, Section 32, John Doerr, Spark Capital, Felicis Ventures, Sound Ventures, Arrowmark, and previous investors.

Press Inquiries [emailprotected]

Related Links https://www.trialspark.com

SOURCE TrialSpark

https://www.trialspark.com

See the article here:
TrialSpark licenses sprifermin, an investigational first-in-class disease modifying treatment for osteoarthritis, from Merck KGaA, Darmstadt, Germany...

Read More...

MS in Stem Cell Biology and Regenerative Medicine

Sunday, November 7th, 2021

Discover the future of medicine

The Master of Science degree program invites you to chart the course for the medicine of the futureregenerative medicine. This is one of the first masters programs in stem cell biology and regenerative medicine in the United States.

Our one-year program offers courses in cutting-edge biomedical science, including developmental biology, human embryology, regenerative medicine, and the translational and therapeutic aspects of stem cell technology. The program also provides practical hands-on laboratory experience with the growth and differentiation of stem cells. Although not required, students are encouraged to engage in laboratory research during the year, with one of the 80+ lab groups that constitute USC Stem Cell. At the completion of the first year, students may informally continue to conduct research in their labs after receiving the MS diploma, or can petition to continue research with a guided and structured second research year culminating in a capstone thesis project.

After completing this program, you will be poised to apply to medical or PhD programs, enter the growing stem cell pharmaceutical domain, or engage in other academic, clinical or business efforts. You will possess a unique understanding of how the bodys own developmental and repair mechanisms can restore damaged cells, tissues and organsproviding new opportunities to treat conditions ranging from blindness to cancer, from organ failure to HIV/AIDS.

To apply, visit gradadm.usc.edu.

Please note that the application portal for Fall 2022 will open October 15th, 2021. The deadline to apply will be April 1st, 2022.

For questions, e-mail us at scrm@usc.edu or call (323) 865 1266.

Read more from the original source:
MS in Stem Cell Biology and Regenerative Medicine

Read More...

Stem Cells Applications in Regenerative Medicine and …

Sunday, November 7th, 2021

Int J Cell Biol. 2016; 2016: 6940283.

Department of Biological Sciences, Indian Institute of Science Education and Research (IISER), Bhopal, Madhya Pradesh 462066, India

Department of Biological Sciences, Indian Institute of Science Education and Research (IISER), Bhopal, Madhya Pradesh 462066, India

Academic Editor: Paul J. Higgins

Received 2016 Mar 13; Accepted 2016 Jun 5.

This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Regenerative medicine, the most recent and emerging branch of medical science, deals with functional restoration of tissues or organs for the patient suffering from severe injuries or chronic disease. The spectacular progress in the field of stem cell research has laid the foundation for cell based therapies of disease which cannot be cured by conventional medicines. The indefinite self-renewal and potential to differentiate into other types of cells represent stem cells as frontiers of regenerative medicine. The transdifferentiating potential of stem cells varies with source and according to that regenerative applications also change. Advancements in gene editing and tissue engineering technology have endorsed the ex vivo remodelling of stem cells grown into 3D organoids and tissue structures for personalized applications. This review outlines the most recent advancement in transplantation and tissue engineering technologies of ESCs, TSPSCs, MSCs, UCSCs, BMSCs, and iPSCs in regenerative medicine. Additionally, this review also discusses stem cells regenerative application in wildlife conservation.

Regenerative medicine, the most recent and emerging branch of medical science, deals with functional restoration of specific tissue and/or organ of the patients suffering with severe injuries or chronic disease conditions, in the state where bodies own regenerative responses do not suffice [1]. In the present scenario donated tissues and organs cannot meet the transplantation demands of aged and diseased populations that have driven the thrust for search for the alternatives. Stem cells are endorsed with indefinite cell division potential, can transdifferentiate into other types of cells, and have emerged as frontline regenerative medicine source in recent time, for reparation of tissues and organs anomalies occurring due to congenital defects, disease, and age associated effects [1]. Stem cells pave foundation for all tissue and organ system of the body and mediates diverse role in disease progression, development, and tissue repair processes in host. On the basis of transdifferentiation potential, stem cells are of four types, that is, (1) unipotent, (2) multipotent, (3) pluripotent, and (4) totipotent [2]. Zygote, the only totipotent stem cell in human body, can give rise to whole organism through the process of transdifferentiation, while cells from inner cells mass (ICM) of embryo are pluripotent in their nature and can differentiate into cells representing three germ layers but do not differentiate into cells of extraembryonic tissue [2]. Stemness and transdifferentiation potential of the embryonic, extraembryonic, fetal, or adult stem cells depend on functional status of pluripotency factors like OCT4, cMYC, KLF44, NANOG, SOX2, and so forth [35]. Ectopic expression or functional restoration of endogenous pluripotency factors epigenetically transforms terminally differentiated cells into ESCs-like cells [3], known as induced pluripotent stem cells (iPSCs) [3, 4]. On the basis of regenerative applications, stem cells can be categorized as embryonic stem cells (ESCs), tissue specific progenitor stem cells (TSPSCs), mesenchymal stem cells (MSCs), umbilical cord stem cells (UCSCs), bone marrow stem cells (BMSCs), and iPSCs (; ). The transplantation of stem cells can be autologous, allogenic, and syngeneic for induction of tissue regeneration and immunolysis of pathogen or malignant cells. For avoiding the consequences of host-versus-graft rejections, tissue typing of human leucocyte antigens (HLA) for tissue and organ transplant as well as use of immune suppressant is recommended [6]. Stem cells express major histocompatibility complex (MHC) receptor in low and secret chemokine that recruitment of endothelial and immune cells is enabling tissue tolerance at graft site [6]. The current stem cell regenerative medicine approaches are founded onto tissue engineering technologies that combine the principles of cell transplantation, material science, and microengineering for development of organoid; those can be used for physiological restoration of damaged tissue and organs. The tissue engineering technology generates nascent tissue on biodegradable 3D-scaffolds [7, 8]. The ideal scaffolds support cell adhesion and ingrowths, mimic mechanics of target tissue, support angiogenesis and neovascularisation for appropriate tissue perfusion, and, being nonimmunogenic to host, do not require systemic immune suppressant [9]. Stem cells number in tissue transplant impacts upon regenerative outcome [10]; in that case prior ex vivo expansion of transplantable stem cells is required [11]. For successful regenerative outcomes, transplanted stem cells must survive, proliferate, and differentiate in site specific manner and integrate into host circulatory system [12]. This review provides framework of most recent (; Figures ) advancement in transplantation and tissue engineering technologies of ESCs, TSPSCs, MSCs, UCSCs, BMSCs, and iPSCs in regenerative medicine. Additionally, this review also discusses stem cells as the tool of regenerative applications in wildlife conservation.

Promises of stem cells in regenerative medicine: the six classes of stem cells, that is, embryonic stem cells (ESCs), tissue specific progenitor stem cells (TSPSCs), mesenchymal stem cells (MSCs), umbilical cord stem cells (UCSCs), bone marrow stem cells (BMSCs), and induced pluripotent stem cells (iPSCs), have many promises in regenerative medicine and disease therapeutics.

ESCs in regenerative medicine: ESCs, sourced from ICM of gastrula, have tremendous promises in regenerative medicine. These cells can differentiate into more than 200 types of cells representing three germ layers. With defined culture conditions, ESCs can be transformed into hepatocytes, retinal ganglion cells, chondrocytes, pancreatic progenitor cells, cone cells, cardiomyocytes, pacemaker cells, eggs, and sperms which can be used in regeneration of tissue and treatment of disease in tissue specific manner.

TSPSCs in regenerative medicine: tissue specific stem and progenitor cells have potential to differentiate into other cells of the tissue. Characteristically inner ear stem cells can be transformed into auditory hair cells, skin progenitors into vascular smooth muscle cells, mesoangioblasts into tibialis anterior muscles, and dental pulp stem cells into serotonin cells. The 3D-culture of TSPSCs in complex biomaterial gives rise to tissue organoids, such as pancreatic organoid from pancreatic progenitor, intestinal tissue organoids from intestinal progenitor cells, and fallopian tube organoids from fallopian tube epithelial cells. Transplantation of TSPSCs regenerates targets tissue such as regeneration of tibialis muscles from mesoangioblasts, cardiac tissue from AdSCs, and corneal tissue from limbal stem cells. Cell growth and transformation factors secreted by TSPSCs can change cells fate to become other types of cell, such that SSCs coculture with skin, prostate, and intestine mesenchyme transforms these cells from MSCs into epithelial cells fate.

MSCs in regenerative medicine: mesenchymal stem cells are CD73+, CD90+, CD105+, CD34, CD45, CD11b, CD14, CD19, and CD79a cells, also known as stromal cells. These bodily MSCs represented here do not account for MSCs of bone marrow and umbilical cord. Upon transplantation and transdifferentiation these bodily MSCs regenerate into cartilage, bones, and muscles tissue. Heart scar formed after heart attack and liver cirrhosis can be treated from MSCs. ECM coating provides the niche environment for MSCs to regenerate into hair follicle, stimulating hair growth.

UCSCs in regenerative medicine: umbilical cord, the readily available source of stem cells, has emerged as futuristic source for personalized stem cell therapy. Transplantation of UCSCs to Krabbe's disease patients regenerates myelin tissue and recovers neuroblastoma patients through restoring tissue homeostasis. The UCSCs organoids are readily available tissue source for treatment of neurodegenerative disease. Peritoneal fibrosis caused by long term dialysis, tendon tissue degeneration, and defective hyaline cartilage can be regenerated by UCSCs. Intravenous injection of UCSCs enables treatment of diabetes, spinal myelitis, systemic lupus erythematosus, Hodgkin's lymphoma, and congenital neuropathies. Cord blood stem cells banking avails long lasting source of stem cells for personalized therapy and regenerative medicine.

BMSCs in regenerative medicine: bone marrow, the soft sponge bone tissue that consisted of stromal, hematopoietic, and mesenchymal and progenitor stem cells, is responsible for blood formation. Even halo-HLA matched BMSCs can cure from disease and regenerate tissue. BMSCs can regenerate craniofacial tissue, brain tissue, diaphragm tissue, and liver tissue and restore erectile function and transdifferentiation monocytes. These multipotent stem cells can cure host from cancer and infection of HIV and HCV.

iPSCs in regenerative medicine: using the edge of iPSCs technology, skin fibroblasts and other adult tissues derived, terminally differentiated cells can be transformed into ESCs-like cells. It is possible that adult cells can be transformed into cells of distinct lineages bypassing the phase of pluripotency. The tissue specific defined culture can transform skin cells to become trophoblast, heart valve cells, photoreceptor cells, immune cells, melanocytes, and so forth. ECM complexation with iPSCs enables generation of tissue organoids for lung, kidney, brain, and other organs of the body. Similar to ESCs, iPSCs also can be transformed into cells representing three germ layers such as pacemaker cells and serotonin cells.

Stem cells in wildlife conservation: tissue biopsies obtained from dead and live wild animals can be either cryopreserved or transdifferentiated to other types of cells, through culture in defined culture medium or in vivo maturation. Stem cells and adult tissue derived iPSCs have great potential of regenerative medicine and disease therapeutics. Gonadal tissue procured from dead wild animals can be matured, ex vivo and in vivo for generation of sperm and egg, which can be used for assistive reproductive technology oriented captive breeding of wild animals or even for resurrection of wildlife.

Application of stem cells in regenerative medicine: stem cells (ESCs, TSPSCs, MSCs, UCSCs, BMSCs, and iPSCs) have diverse applications in tissue regeneration and disease therapeutics.

For the first time in 1998, Thomson isolated human ESCs (hESCs) [13]. ESCs are pluripotent in their nature and can give rise to more than 200 types of cells and promises for the treatment of any kinds of disease [13]. The pluripotency fate of ESCs is governed by functional dynamics of transcription factors OCT4, SOX2, NANOG, and so forth, which are termed as pluripotency factors. The two alleles of the OCT4 are held apart in pluripotency state in ESCs; phase through homologues pairing during embryogenesis and transdifferentiation processes [14] has been considered as critical regulatory switch for lineage commitment of ESCs. The diverse lineage commitment potential represents ESCs as ideal model for regenerative therapeutics of disease and tissue anomalies. This section of review on ESCs discusses transplantation and transdifferentiation of ESCs into retinal ganglion, hepatocytes, cardiomyocytes, pancreatic progenitors, chondrocytes, cones, egg sperm, and pacemaker cells (; ). Infection, cancer treatment, and accidents can cause spinal cord injuries (SCIs). The transplantation of hESCs to paraplegic or quadriplegic SCI patients improves body control, balance, sensation, and limbal movements [15], where transplanted stem cells do homing to injury sites. By birth, humans have fixed numbers of cone cells; degeneration of retinal pigment epithelium (RPE) of macula in central retina causes age-related macular degeneration (ARMD). The genomic incorporation of COCO gene (expressed during embryogenesis) in the developing embryo leads lineage commitment of ESCs into cone cells, through suppression of TGF, BMP, and Wnt signalling pathways. Transplantation of these cone cells to eye recovers individual from ARMD phenomenon, where transplanted cone cells migrate and form sheet-like structure in host retina [16]. However, establishment of missing neuronal connection of retinal ganglion cells (RGCs), cones, and PRE is the most challenging aspect of ARMD therapeutics. Recently, Donald Z Jacks group at John Hopkins University School of Medicine has generated RGCs from CRISPER-Cas9-m-Cherry reporter ESCs [17]. During ESCs transdifferentiation process, CRIPER-Cas9 directs the knock-in of m-Cherry reporter into 3UTR of BRN3B gene, which is specifically expressed in RGCs and can be used for purification of generated RGCs from other cells [17]. Furthermore, incorporation of forskolin in transdifferentiation regime boosts generation of RGCs. Coaxing of these RGCs into biomaterial scaffolds directs axonal differentiation of RGCs. Further modification in RGCs generation regime and composition of biomaterial scaffolds might enable restoration of vision for ARMD and glaucoma patients [17]. Globally, especially in India, cardiovascular problems are a more common cause of human death, where biomedical therapeutics require immediate restoration of heart functions for the very survival of the patient. Regeneration of cardiac tissue can be achieved by transplantation of cardiomyocytes, ESCs-derived cardiovascular progenitors, and bone marrow derived mononuclear cells (BMDMNCs); however healing by cardiomyocytes and progenitor cells is superior to BMDMNCs but mature cardiomyocytes have higher tissue healing potential, suppress heart arrhythmias, couple electromagnetically into hearts functions, and provide mechanical and electrical repair without any associated tumorigenic effects [18, 19]. Like CM differentiation, ESCs derived liver stem cells can be transformed into Cytp450-hepatocytes, mediating chemical modification and catabolism of toxic xenobiotic drugs [20]. Even today, availability and variability of functional hepatocytes are a major a challenge for testing drug toxicity [20]. Stimulation of ESCs and ex vivo VitK12 and lithocholic acid (a by-product of intestinal flora regulating drug metabolism during infancy) activates pregnane X receptor (PXR), CYP3A4, and CYP2C9, which leads to differentiation of ESCs into hepatocytes; those are functionally similar to primary hepatocytes, for their ability to produce albumin and apolipoprotein B100 [20]. These hepatocytes are excellent source for the endpoint screening of drugs for accurate prediction of clinical outcomes [20]. Generation of hepatic cells from ESCs can be achieved in multiple ways, as serum-free differentiation [21], chemical approaches [20, 22], and genetic transformation [23, 24]. These ESCs-derived hepatocytes are long lasting source for treatment of liver injuries and high throughput screening of drugs [20, 23, 24]. Transplantation of the inert biomaterial encapsulated hESCs-derived pancreatic progenitors (CD24+, CD49+, and CD133+) differentiates into -cells, minimizing high fat diet induced glycemic and obesity effects in mice [25] (). Addition of antidiabetic drugs into transdifferentiation regime can boost ESCs conservation into -cells [25], which theoretically can cure T2DM permanently [25]. ESCs can be differentiated directly into insulin secreting -cells (marked with GLUT2, INS1, GCK, and PDX1) which can be achieved through PDX1 mediated epigenetic reprogramming [26]. Globally, osteoarthritis affects millions of people and occurs when cartilage at joints wears away, causing stiffness of the joints. The available therapeutics for arthritis relieve symptoms but do not initiate reverse generation of cartilage. For young individuals and athletes replacement of joints is not feasible like old populations; in that case transplantation of stem cells represents an alternative for healing cartilage injuries [27]. Chondrocytes, the cartilage forming cells derived from hESC, embedded in fibrin gel effectively heal defective cartilage within 12 weeks, when transplanted to focal cartilage defects of knee joints in mice without any negative effect [27]. Transplanted chondrocytes form cell aggregates, positive for SOX9 and collagen II, and defined chondrocytes are active for more than 12wks at transplantation site, advocating clinical suitability of chondrocytes for treatment of cartilage lesions [27]. The integrity of ESCs to integrate and differentiate into electrophysiologically active cells provides a means for natural regulation of heart rhythm as biological pacemaker. Coaxing of ESCs into inert biomaterial as well as propagation in defined culture conditions leads to transdifferentiation of ESCs to become sinoatrial node (SAN) pacemaker cells (PCs) [28]. Genomic incorporation TBox3 into ESCs ex vivo leads to generation of PCs-like cells; those express activated leukocyte cells adhesion molecules (ALCAM) and exhibit similarity to PCs for gene expression and immune functions [28]. Transplantation of PCs can restore pacemaker functions of the ailing heart [28]. In summary, ESCs can be transdifferentiated into any kinds of cells representing three germ layers of the body, being most promising source of regenerative medicine for tissue regeneration and disease therapy (). Ethical concerns limit the applications of ESCs, where set guidelines need to be followed; in that case TSPSCs, MSCs, UCSCs, BMSCs, and iPSCs can be explored as alternatives.

TSPSCs maintain tissue homeostasis through continuous cell division, but, unlike ESCs, TSPSCs retain stem cells plasticity and differentiation in tissue specific manner, giving rise to few types of cells (). The number of TSPSCs population to total cells population is too low; in that case their harvesting as well as in vitro manipulation is really a tricky task [29], to explore them for therapeutic scale. Human body has foundation from various types of TSPSCs; discussing the therapeutic application for all types is not feasible. This section of review discusses therapeutic application of pancreatic progenitor cells (PPCs), dental pulp stem cells (DPSCs), inner ear stem cells (IESCs), intestinal progenitor cells (IPCs), limbal progenitor stem cells (LPSCs), epithelial progenitor stem cells (EPSCs), mesoangioblasts (MABs), spermatogonial stem cells (SSCs), the skin derived precursors (SKPs), and adipose derived stem cells (AdSCs) (; ). During embryogenesis PPCs give rise to insulin-producing -cells. The differentiation of PPCs to become -cells is negatively regulated by insulin [30]. PPCs require active FGF and Notch signalling; growing more rapidly in community than in single cell populations advocates the functional importance of niche effect in self-renewal and transdifferentiation processes. In 3D-scaffold culture system, mice embryo derived PPCs grow into hollow organoid spheres; those finally differentiate into insulin-producing -cell clusters [29]. The DSPSCs, responsible for maintenance of teeth health status, can be sourced from apical papilla, deciduous teeth, dental follicle, and periodontal ligaments, have emerged as regenerative medicine candidate, and might be explored for treatment of various kinds of disease including restoration neurogenic functions in teeth [31, 32]. Expansion of DSPSCs in chemically defined neuronal culture medium transforms them into a mixed population of cholinergic, GABAergic, and glutaminergic neurons; those are known to respond towards acetylcholine, GABA, and glutamine stimulations in vivo. These transformed neuronal cells express nestin, glial fibrillary acidic protein (GFAP), III-tubulin, and voltage gated L-type Ca2+ channels [32]. However, absence of Na+ and K+ channels does not support spontaneous action potential generation, necessary for response generation against environmental stimulus. All together, these primordial neuronal stem cells have possible therapeutic potential for treatment of neurodental problems [32]. Sometimes, brain tumor chemotherapy can cause neurodegeneration mediated cognitive impairment, a condition known as chemobrain [33]. The intrahippocampal transplantation of human derived neuronal stem cells to cyclophosphamide behavioural decremented mice restores cognitive functions in a month time. Here the transplanted stem cells differentiate into neuronal and astroglial lineage, reduce neuroinflammation, and restore microglial functions [33]. Furthermore, transplantation of stem cells, followed by chemotherapy, directs pyramidal and granule-cell neurons of the gyrus and CA1 subfields of hippocampus which leads to reduction in spine and dendritic cell density in the brain. These findings suggest that transplantation of stem cells to cranium restores cognitive functions of the chemobrain [33]. The hair cells of the auditory system produced during development are not postmitotic; loss of hair cells cannot be replaced by inner ear stem cells, due to active state of the Notch signalling [34]. Stimulation of inner ear progenitors with -secretase inhibitor ({"type":"entrez-nucleotide","attrs":{"text":"LY411575","term_id":"1257853995","term_text":"LY411575"}}LY411575) abrogates Notch signalling through activation of transcription factor atonal homologue 1 (Atoh1) and directs transdifferentiation of progenitors into cochlear hair cells [34]. Transplantation of in vitro generated hair cells restores acoustic functions in mice, which can be the potential regenerative medicine candidates for the treatment of deafness [34]. Generation of the hair cells also can be achieved through overexpression of -catenin and Atoh1 in Lrg5+ cells in vivo [35]. Similar to ear progenitors, intestine of the digestive tract also has its own tissue specific progenitor stem cells, mediating regeneration of the intestinal tissue [34, 36]. Dysregulation of the common stem cells signalling pathways, Notch/BMP/TGF-/Wnt, in the intestinal tissue leads to disease. Information on these signalling pathways [37] is critically important in designing therapeutics. Coaxing of the intestinal tissue specific progenitors with immune cells (macrophages), connective tissue cells (myofibroblasts), and probiotic bacteria into 3D-scaffolds of inert biomaterial, crafting biological environment, is suitable for differentiation of progenitors to occupy the crypt-villi structures into these scaffolds [36]. Omental implementation of these crypt-villi structures to dogs enhances intestinal mucosa through regeneration of goblet cells containing intestinal tissue [36]. These intestinal scaffolds are close approach for generation of implantable intestinal tissue, divested by infection, trauma, cancer, necrotizing enterocolitis (NEC), and so forth [36]. In vitro culture conditions cause differentiation of intestinal stem cells to become other types of cells, whereas incorporation of valproic acid and CHIR-99021 in culture conditions avoids differentiation of intestinal stem cells, enabling generation of indefinite pool of stem cells to be used for regenerative applications [38]. The limbal stem cells of the basal limbal epithelium, marked with ABCB5, are essential for regeneration and maintenance of corneal tissue [39]. Functional status of ABCB5 is critical for survival and functional integrity of limbal stem cells, protecting them from apoptotic cell death [39]. Limbal stem cells deficiency leads to replacement of corneal epithelium with visually dead conjunctival tissue, which can be contributed by burns, inflammation, and genetic factors [40]. Transplanted human cornea stem cells to mice regrown into fully functional human cornea, possibly supported by blood eye barrier phenomena, can be used for treatment of eye diseases, where regeneration of corneal tissue is critically required for vision restoration [39]. Muscle degenerative disease like duchenne muscular dystrophy (DMD) can cause extensive thrashing of muscle tissue, where tissue engineering technology can be deployed for functional restoration of tissue through regeneration [41]. Encapsulation of mouse or human derived MABs (engineered to express placental derived growth factor (PDGF)) into polyethylene glycol (PEG) fibrinogen hydrogel and their transplantation beneath the skin at ablated tibialis anterior form artificial muscles, which are functionally similar to those of normal tibialis anterior muscles [41]. The PDGF attracts various cell types of vasculogenic and neurogenic potential to the site of transplantation, supporting transdifferentiation of mesoangioblasts to become muscle fibrils [41]. The therapeutic application of MABs in skeletal muscle regeneration and other therapeutic outcomes has been reviewed by others [42]. One of the most important tissue specific stem cells, the male germline stem cells or spermatogonial stem cells (SSCs), produces spermatogenic lineage through mesenchymal and epithets cells [43] which itself creates niche effect on other cells. In vivo transplantation of SSCs with prostate, skin, and uterine mesenchyme leads to differentiation of these cells to become epithelia of the tissue of origin [43]. These newly formed tissues exhibit all physical and physiological characteristics of prostate and skin and the physical characteristics of prostate, skin, and uterus, express tissue specific markers, and suggest that factors secreted from SSCs lead to lineage conservation which defines the importance of niche effect in regenerative medicine [43]. According to an estimate, more than 100 million people are suffering from the condition of diabetic retinopathy, a progressive dropout of vascularisation in retina that leads to loss of vision [44]. The intravitreal injection of adipose derived stem cells (AdSCs) to the eye restores microvascular capillary bed in mice. The AdSCs from healthy donor produce higher amounts of vasoprotective factors compared to glycemic mice, enabling superior vascularisation [44]. However use of AdSCs for disease therapeutics needs further standardization for cell counts in dose of transplant and monitoring of therapeutic outcomes at population scale [44]. Apart from AdSCs, other kinds of stem cells also have therapeutic potential in regenerative medicine for treatment of eye defects, which has been reviewed by others [45]. Fallopian tubes, connecting ovaries to uterus, are the sites where fertilization of the egg takes place. Infection in fallopian tubes can lead to inflammation, tissue scarring, and closure of the fallopian tube which often leads to infertility and ectopic pregnancies. Fallopian is also the site where onset of ovarian cancer takes place. The studies on origin and etiology of ovarian cancer are restricted due to lack of technical advancement for culture of epithelial cells. The in vitro 3D organoid culture of clinically obtained fallopian tube epithelial cells retains their tissue specificity, keeps cells alive, which differentiate into typical ciliated and secretory cells of fallopian tube, and advocates that ectopic examination of fallopian tube in organoid culture settings might be the ideal approach for screening of cancer [46]. The sustained growth and differentiation of fallopian TSPSCs into fallopian tube organoid depend both on the active state of the Wnt and on paracrine Notch signalling [46]. Similar to fallopian tube stem cells, subcutaneous visceral tissue specific cardiac adipose (CA) derived stem cells (AdSCs) have the potential of differentiation into cardiovascular tissue [47]. Systemic infusion of CA-AdSCs into ischemic myocardium of mice regenerates heart tissue and improves cardiac function through differentiation to endothelial cells, vascular smooth cells, and cardiomyocytes and vascular smooth cells. The differentiation and heart regeneration potential of CA-AdSCs are higher than AdSCs [48], representing CA-AdSCs as potent regenerative medicine candidates for myocardial ischemic therapy [47]. The skin derived precursors (SKPs), the progenitors of dermal papilla/hair/hair sheath, give rise to multiple tissues of mesodermal and/or ectodermal origin such as neurons, Schwann cells, adipocytes, chondrocytes, and vascular smooth muscle cells (VSMCs). VSMCs mediate wound healing and angiogenesis process can be derived from human foreskin progenitor SKPs, suggesting that SKPs derived VSMCs are potential regenerative medicine candidates for wound healing and vasculature injuries treatments [49]. In summary, TSPSCs are potentiated with tissue regeneration, where advancement in organoid culture (; ) technologies defines the importance of niche effect in tissue regeneration and therapeutic outcomes of ex vivo expanded stem cells.

MSCs, the multilineage stem cells, differentiate only to tissue of mesodermal origin, which includes tendons, bone, cartilage, ligaments, muscles, and neurons [50]. MSCs are the cells which express combination of markers: CD73+, CD90+, CD105+, CD11b, CD14, CD19, CD34, CD45, CD79a, and HLA-DR, reviewed elsewhere [50]. The application of MSCs in regenerative medicine can be generalized from ongoing clinical trials, phasing through different state of completions, reviewed elsewhere [90]. This section of review outlines the most recent representative applications of MSCs (; ). The anatomical and physiological characteristics of both donor and receiver have equal impact on therapeutic outcomes. The bone marrow derived MSCs (BMDMSCs) from baboon are morphologically and phenotypically similar to those of bladder stem cells and can be used in regeneration of bladder tissue. The BMDMSCs (CD105+, CD73+, CD34, and CD45), expressing GFP reporter, coaxed with small intestinal submucosa (SIS) scaffolds, augment healing of degenerated bladder tissue within 10wks of the transplantation [51]. The combinatorial CD characterized MACs are functionally active at transplantation site, which suggests that CD characterization of donor MSCs yields superior regenerative outcomes [51]. MSCs also have potential to regenerate liver tissue and treat liver cirrhosis, reviewed elsewhere [91]. The regenerative medicinal application of MSCs utilizes cells in two formats as direct transplantation or first transdifferentiation and then transplantation; ex vivo transdifferentiation of MSCs deploys retroviral delivery system that can cause oncogenic effect on cells. Nonviral, NanoScript technology, comprising utility of transcription factors (TFs) functionalized gold nanoparticles, can target specific regulatory site in the genome effectively and direct differentiation of MSCs into another cell fate, depending on regime of TFs. For example, myogenic regulatory factor containing NanoScript-MRF differentiates the adipose tissue derived MSCs into muscle cells [92]. The multipotency characteristics represent MSCs as promising candidate for obtaining stable tissue constructs through coaxed 3D organoid culture; however heterogeneous distribution of MSCs slows down cell proliferation, rendering therapeutic applications of MSCs. Adopting two-step culture system for MSCs can yield homogeneous distribution of MSCs in biomaterial scaffolds. For example, fetal-MSCs coaxed in biomaterial when cultured first in rotating bioreactor followed with static culture lead to homogeneous distribution of MSCs in ECM components [7]. Occurrence of dental carries, periodontal disease, and tooth injury can impact individual's health, where bioengineering of teeth can be the alternative option. Coaxing of epithelial-MSCs with dental stem cells into synthetic polymer gives rise to mature teeth unit, which consisted of mature teeth and oral tissue, offering multiple regenerative therapeutics, reviewed elsewhere [52]. Like the tooth decay, both human and animals are prone to orthopedic injuries, affecting bones, joint, tendon, muscles, cartilage, and so forth. Although natural healing potential of bone is sufficient to heal the common injuries, severe trauma and tumor-recession can abrogate germinal potential of bone-forming stem cells. In vitro chondrogenic, osteogenic, and adipogenic potential of MSCs advocates therapeutic applications of MSCs in orthopedic injuries [53]. Seeding of MSCs, coaxed into biomaterial scaffolds, at defective bone tissue, regenerates defective bone tissues, within fourwks of transplantation; by the end of 32wks newly formed tissues integrate into old bone [54]. Osteoblasts, the bone-forming cells, have lesser actin cytoskeleton compared to adipocytes and MSCs. Treatment of MSCs with cytochalasin-D causes rapid transportation of G-actin, leading to osteogenic transformation of MSCs. Furthermore, injection of cytochalasin-D to mice tibia also promotes bone formation within a wk time frame [55]. The bone formation processes in mice, dog, and human are fundamentally similar, so outcomes of research on mice and dogs can be directional for regenerative application to human. Injection of MSCs to femur head of Legg-Calve-Perthes suffering dog heals the bone very fast and reduces the injury associated pain [55]. Degeneration of skeletal muscle and muscle cramps are very common to sledge dogs, animals, and individuals involved in adventurous athletics activities. Direct injection of adipose tissue derived MSCs to tear-site of semitendinosus muscle in dogs heals injuries much faster than traditional therapies [56]. Damage effect treatment for heart muscle regeneration is much more complex than regeneration of skeletal muscles, which needs high grade fine-tuned coordination of neurons with muscles. Coaxing of MSCs into alginate gel increases cell retention time that leads to releasing of tissue repairing factors in controlled manner. Transplantation of alginate encapsulated cells to mice heart reduces scar size and increases vascularisation, which leads to restoration of heart functions. Furthermore, transplanted MSCs face host inhospitable inflammatory immune responses and other mechanical forces at transplantation site, where encapsulation of cells keeps them away from all sorts of mechanical forces and enables sensing of host tissue microenvironment, and respond accordingly [57]. Ageing, disease, and medicine consumption can cause hair loss, known as alopecia. Although alopecia has no life threatening effects, emotional catchments can lead to psychological disturbance. The available treatments for alopecia include hair transplantation and use of drugs, where drugs are expensive to afford and generation of new hair follicle is challenging. Dermal papillary cells (DPCs), the specialized MSCs localized in hair follicle, are responsible for morphogenesis of hair follicle and hair cycling. The layer-by-layer coating of DPCs, called GAG coating, consists of coating of geletin as outer layer, middle layer of fibroblast growth factor 2 (FGF2) loaded alginate, and innermost layer of geletin. GAG coating creates tissue microenvironment for DPCs that can sustain immunological and mechanical obstacles, supporting generation of hair follicle. Transplantation of GAG-coated DPCs leads to abundant hair growth and maturation of hair follicle, where GAG coating serves as ECM, enhancing intrinsic therapeutic potential of DPCs [58]. During infection, the inflammatory cytokines secreted from host immune cells attract MSCs to the site of inflammation, which modulates inflammatory responses, representing MSCs as key candidate of regenerative medicine for infectious disease therapeutics. Coculture of macrophages (M) and adipose derived MSCs from Leishmania major (LM) susceptible and resistant mice demonstrates that AD-MSCs educate M against LM infection, differentially inducing M1 and M2 phenotype that represents AD-MSC as therapeutic agent for leishmanial therapy [93]. In summary, the multilineage differentiation potential of MSCs, as well as adoption of next-generation organoid culture system, avails MSCs as ideal regenerative medicine candidate.

Umbilical cord, generally thrown at the time of child birth, is the best known source for stem cells, procured in noninvasive manner, having lesser ethical constraints than ESCs. Umbilical cord is rich source of hematopoietic stem cells (HSCs) and MSCs, which possess enormous regeneration potential [94] (; ). The HSCs of cord blood are responsible for constant renewal of all types of blood cells and protective immune cells. The proliferation of HSCs is regulated by Musashi-2 protein mediated attenuation of Aryl hydrocarbon receptor (AHR) signalling in stem cells [95]. UCSCs can be cryopreserved at stem cells banks (; ), in operation by both private and public sector organization. Public stem cells banks operate on donation formats and perform rigorous screening for HLA typing and donated UCSCs remain available to anyone in need, whereas private stem cell banks operation is more personalized, availing cells according to donor consent. Stem cell banking is not so common, even in developed countries. Survey studies find that educated women are more eager to donate UCSCs, but willingness for donation decreases with subsequent deliveries, due to associated cost and safety concerns for preservation [96]. FDA has approved five HSCs for treatment of blood and other immunological complications [97]. The amniotic fluid, drawn during pregnancy for standard diagnostic purposes, is generally discarded without considering its vasculogenic potential. UCSCs are the best alternatives for those patients who lack donors with fully matched HLA typing for peripheral blood and PBMCs and bone marrow [98]. One major issue with UCSCs is number of cells in transplant, fewer cells in transplant require more time for engraftment to mature, and there are also risks of infection and mortality; in that case ex vivo propagation of UCSCs can meet the demand of desired outcomes. There are diverse protocols, available for ex vivo expansion of UCSCs, reviewed elsewhere [99]. Amniotic fluid stem cells (AFSCs), coaxed to fibrin (required for blood clotting, ECM interactions, wound healing, and angiogenesis) hydrogel and PEG supplemented with vascular endothelial growth factor (VEGF), give rise to vascularised tissue, when grafted to mice, suggesting that organoid cultures of UCSCs have promise for generation of biocompatible tissue patches, for treating infants born with congenital heart defects [59]. Retroviral integration of OCT4, KLF4, cMYC, and SOX2 transforms AFSCs into pluripotency stem cells known as AFiPSCs which can be directed to differentiate into extraembryonic trophoblast by BMP2 and BMP4 stimulation, which can be used for regeneration of placental tissues [60]. Wharton's jelly (WJ), the gelatinous substance inside umbilical cord, is rich in mucopolysaccharides, fibroblast, macrophages, and stem cells. The stem cells from UCB and WJ can be transdifferentiated into -cells. Homogeneous nature of WJ-SCs enables better differentiation into -cells; transplantation of these cells to streptozotocin induced diabetic mice efficiently brings glucose level to normal [7]. Easy access and expansion potential and plasticity to differentiate into multiple cell lineages represent WJ as an ideal candidate for regenerative medicine but cells viability changes with passages with maximum viable population at 5th-6th passages. So it is suggested to perform controlled expansion of WJ-MSCS for desired regenerative outcomes [9]. Study suggests that CD34+ expression leads to the best regenerative outcomes, with less chance of host-versus-graft rejection. In vitro expansion of UCSCs, in presence of StemRegenin-1 (SR-1), conditionally expands CD34+ cells [61]. In type I diabetic mellitus (T1DM), T-cell mediated autoimmune destruction of pancreatic -cells occurs, which has been considered as tough to treat. Transplantation of WJ-SCs to recent onset-T1DM patients restores pancreatic function, suggesting that WJ-MSCs are effective in regeneration of pancreatic tissue anomalies [62]. WJ-MSCs also have therapeutic importance for treatment of T2DM. A non-placebo controlled phase I/II clinical trial demonstrates that intravenous and intrapancreatic endovascular injection of WJ-MSCs to T2DM patients controls fasting glucose and glycated haemoglobin through improvement of -cells functions, evidenced by enhanced c-peptides and reduced inflammatory cytokines (IL-1 and IL-6) and T-cells counts [63]. Like diabetes, systematic lupus erythematosus (SLE) also can be treated with WJ-MSCs transplantation. During progression of SLE host immune system targets its own tissue leading to degeneration of renal, cardiovascular, neuronal, and musculoskeletal tissues. A non-placebo controlled follow-up study on 40 SLE patients demonstrates that intravenous infusion of WJ-MSC improves renal functions and decreases systematic lupus erythematosus disease activity index (SLEDAI) and British Isles Lupus Assessment Group (BILAG), and repeated infusion of WJ-MSCs protects the patient from relapse of the disease [64]. Sometimes, host inflammatory immune responses can be detrimental for HSCs transplantation and blood transfusion procedures. Infusion of WJ-MSC to patients, who had allogenic HSCs transplantation, reduces haemorrhage inflammation (HI) of bladder, suggesting that WJ-MSCs are potential stem cells adjuvant in HSCs transplantation and blood transfusion based therapies [100]. Apart from WJ, umbilical cord perivascular space and cord vein are also rich source for obtaining MSCs. The perivascular MSCs of umbilical cord are more primitive than WJ-MSCs and other MSCs from cord suggest that perivascular MSCs might be used as alternatives for WJ-MSCs for regenerative therapeutics outcome [101]. Based on origin, MSCs exhibit differential in vitro and in vivo properties and advocate functional characterization of MSCs, prior to regenerative applications. Emerging evidence suggests that UCSCs can heal brain injuries, caused by neurodegenerative diseases like Alzheimer's, Krabbe's disease, and so forth. Krabbe's disease, the infantile lysosomal storage disease, occurs due to deficiency of myelin synthesizing enzyme (MSE), affecting brain development and cognitive functions. Progression of neurodegeneration finally leads to death of babies aged two. Investigation shows that healing of peripheral nervous system (PNS) and central nervous system (CNS) tissues with Krabbe's disease can be achieved by allogenic UCSCs. UCSCs transplantation to asymptomatic infants with subsequent monitoring for 46 years reveals that UCSCs recover babies from MSE deficiency, improving myelination and cognitive functions, compared to those of symptomatic babies. The survival rate of transplanted UCSCs in asymptomatic and symptomatic infants was 100% and 43%, respectively, suggesting that early diagnosis and timely treatment are critical for UCSCs acceptance for desired therapeutic outcomes. UCSCs are more primitive than BMSCs, so perfect HLA typing is not critically required, representing UCSCs as an excellent source for treatment of all the diseases involving lysosomal defects, like Krabbe's disease, hurler syndrome, adrenoleukodystrophy (ALD), metachromatic leukodystrophy (MLD), Tay-Sachs disease (TSD), and Sandhoff disease [65]. Brain injuries often lead to cavities formation, which can be treated from neuronal parenchyma, generated ex vivo from UCSCs. Coaxing of UCSCs into human originated biodegradable matrix scaffold and in vitro expansion of cells in defined culture conditions lead to formation of neuronal organoids, within threewks' time frame. These organoids structurally resemble brain tissue and consisted of neuroblasts (GFAP+, Nestin+, and Ki67+) and immature stem cells (OCT4+ and SOX2+). The neuroblasts of these organoids further can be differentiated into mature neurons (MAP2+ and TUJ1+) [66]. Administration of high dose of drugs in divesting neuroblastoma therapeutics requires immediate restoration of hematopoiesis. Although BMSCs had been promising in restoration of hematopoiesis UCSCs are sparely used in clinical settings. A case study demonstrates that neuroblastoma patients who received autologous UCSCs survive without any associated side effects [12]. During radiation therapy of neoplasm, spinal cord myelitis can occur, although occurrence of myelitis is a rare event and usually such neurodegenerative complication of spinal cord occurs 624 years after exposure to radiations. Transplantation of allogenic UC-MSCs in laryngeal patients undergoing radiation therapy restores myelination [102]. For treatment of neurodegenerative disease like Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), traumatic brain injuries (TBI), Parkinson's, SCI, stroke, and so forth, distribution of transplanted UCSCs is critical for therapeutic outcomes. In mice and rat, injection of UCSCs and subsequent MRI scanning show that transplanted UCSCs migrate to CNS and multiple peripheral organs [67]. For immunomodulation of tumor cells disease recovery, transplantation of allogenic DCs is required. The CD11c+DCs, derived from UCB, are morphologically and phenotypically similar to those of peripheral blood derived CTLs-DCs, suggesting that UCB-DCs can be used for personalized medicine of cancer patient, in need for DCs transplantation [103]. Coculture of UCSCs with radiation exposed human lung fibroblast stops their transdifferentiation, which suggests that factors secreted from UCSCs may restore niche identity of fibroblast, if they are transplanted to lung after radiation therapy [104]. Tearing of shoulder cuff tendon can cause severe pain and functional disability, whereas ultrasound guided transplantation of UCB-MSCs in rabbit regenerates subscapularis tendon in fourwks' time frame, suggesting that UCB-MSCs are effective enough to treat tendons injuries when injected to focal points of tear-site [68]. Furthermore, transplantation of UCB-MSCs to chondral cartilage injuries site in pig knee along with HA hydrogel composite regenerates hyaline cartilage [69], suggesting that UCB-MSCs are effective regenerative medicine candidate for treating cartilage and ligament injuries. Physiologically circulatory systems of brain, placenta, and lungs are similar. Infusion of UCB-MSCs to preeclampsia (PE) induced hypertension mice reduces the endotoxic effect, suggesting that UC-MSCs are potential source for treatment of endotoxin induced hypertension during pregnancy, drug abuse, and other kinds of inflammatory shocks [105]. Transplantation of UCSCs to severe congenital neutropenia (SCN) patients restores neutrophils count from donor cells without any side effect, representing UCSCs as potential alternative for SCN therapy, when HLA matched bone marrow donors are not accessible [106]. In clinical settings, the success of myocardial infarction (MI) treatment depends on ageing, systemic inflammation in host, and processing of cells for infusion. Infusion of human hyaluronan hydrogel coaxed UCSCs in pigs induces angiogenesis, decreases scar area, improves cardiac function at preclinical level, and suggests that the same strategy might be effective for human [107]. In stem cells therapeutics, UCSCs transplantation can be either autologous or allogenic. Sometimes, the autologous UCSCs transplants cannot combat over tumor relapse, observed in Hodgkin's lymphoma (HL), which might require second dose transplantation of allogenic stem cells, but efficacy and tolerance of stem cells transplant need to be addressed, where tumor replace occurs. A case study demonstrates that second dose allogenic transplants of UCSCs effective for HL patients, who had heavy dose in prior transplant, increase the long term survival chances by 30% [10]. Patients undergoing long term peritoneal renal dialysis are prone to peritoneal fibrosis and can change peritoneal structure and failure of ultrafiltration processes. The intraperitoneal (IP) injection of WJ-MSCs prevents methylglyoxal induced programmed cell death and peritoneal wall thickening and fibrosis, suggesting that WJ-MSCs are effective in therapeutics of encapsulating peritoneal fibrosis [70]. In summary, UCB-HSCs, WJ-MSCs, perivascular MSCs, and UCB-MSCs have tissue regeneration potential.

Bone marrow found in soft spongy bones is responsible for formation of all peripheral blood and comprises hematopoietic stem cells (producing blood cells) and stromal cells (producing fat, cartilage, and bones) [108] (; ). Visually bone marrow has two types, red marrow (myeloid tissue; producing RBC, platelets, and most of WBC) and yellow marrow (producing fat cells and some WBC) [108]. Imbalance in marrow composition can culminate to the diseased condition. Since 1980, bone marrow transplantation is widely accepted for cancer therapeutics [109]. In order to avoid graft rejection, HLA typing of donors is a must, but completely matched donors are limited to family members, which hampers allogenic transplantation applications. Since matching of all HLA antigens is not critically required, in that case defining the critical antigens for haploidentical allogenic donor for patients, who cannot find fully matched donor, might relieve from donor constraints. Two-step administration of lymphoid and myeloid BMSCs from haploidentical donor to the patients of aplastic anaemia and haematological malignancies reconstructs host immune system and the outcomes are almost similar to fully matched transplants, which recommends that profiling of critically important HLA is sufficient for successful outcomes of BMSCs transplantation. Haploidentical HLA matching protocol is the major process for minorities and others who do not have access to matched donor [71]. Furthermore, antigen profiling is not the sole concern for BMSCs based therapeutics. For example, restriction of HIV1 (human immune deficiency virus) infection is not feasible through BMSCs transplantation because HIV1 infection is mediated through CD4+ receptors, chemokine CXC motif receptor 4 (CXCR4), and chemokine receptor 5 (CCR5) for infecting and propagating into T helper (Th), monocytes, macrophages, and dendritic cells (DCs). Genetic variation in CCR2 and CCR5 receptors is also a contributory factor; mediating protection against infection has been reviewed elsewhere [110]. Engineering of hematopoietic stem and progenitor cells (HSPCs) derived CD4+ cells to express HIV1 antagonistic RNA, specifically designed for targeting HIV1 genome, can restrict HIV1 infection, through immune elimination of latently infected CD4+ cells. A single dose infusion of genetically modified (GM), HIV1 resistant HSPCs can be the alternative of HIV1 retroviral therapy. In the present scenario stem cells source, patient selection, transplantation-conditioning regimen, and postinfusion follow-up studies are the major factors, which can limit application of HIV1 resistant GM-HSPCs (CD4+) cells application in AIDS therapy [72, 73]. Platelets, essential for blood clotting, are formed from megakaryocytes inside the bone marrow [74]. Due to infection, trauma, and cancer, there are chances of bone marrow failure. To an extent, spongy bone marrow microenvironment responsible for lineage commitment can be reconstructed ex vivo [75]. The ex vivo constructed 3D-scaffolds consisted of microtubule and silk sponge, flooded with chemically defined organ culture medium, which mimics bone marrow environment. The coculture of megakaryocytes and embryonic stem cells (ESCs) in this microenvironment leads to generation of functional platelets from megakaryocytes [75]. The ex vivo 3D-scaffolds of bone microenvironment can stride the path for generation of platelets in therapeutic quantities for regenerative medication of burns [75] and blood clotting associated defects. Accidents, traumatic injuries, and brain stroke can deplete neuronal stem cells (NSCs), responsible for generation of neurons, astrocytes, and oligodendrocytes. Brain does not repopulate NSCs and heal traumatic injuries itself and transplantation of BMSCs also can heal neurodegeneration alone. Lipoic acid (LA), a known pharmacological antioxidant compound used in treatment of diabetic and multiple sclerosis neuropathy when combined with BMSCs, induces neovascularisation at focal cerebral injuries, within 8wks of transplantation. Vascularisation further attracts microglia and induces their colonization into scaffold, which leads to differentiation of BMSCs to become brain tissue, within 16wks of transplantation. In this approach, healing of tissue directly depends on number of BMSCs in transplantation dose [76]. Dental caries and periodontal disease are common craniofacial disease, often requiring jaw bone reconstruction after removal of the teeth. Traditional therapy focuses on functional and structural restoration of oral tissue, bone, and teeth rather than biological restoration, but BMSCs based therapies promise for regeneration of craniofacial bone defects, enabling replacement of missing teeth in restored bones with dental implants. Bone marrow derived CD14+ and CD90+ stem and progenitor cells, termed as tissue repair cells (TRC), accelerate alveolar bone regeneration and reconstruction of jaw bone when transplanted in damaged craniofacial tissue, earlier to oral implants. Hence, TRC therapy reduces the need of secondary bone grafts, best suited for severe defects in oral bone, skin, and gum, resulting from trauma, disease, or birth defects [77]. Overall, HSCs have great value in regenerative medicine, where stem cells transplantation strategies explore importance of niche in tissue regeneration. Prior to transplantation of BMSCs, clearance of original niche from target tissue is necessary for generation of organoid and organs without host-versus-graft rejection events. Some genetic defects can lead to disorganization of niche, leading to developmental errors. Complementation with human blastocyst derived primary cells can restore niche function of pancreas in pigs and rats, which defines the concept for generation of clinical grade human pancreas in mice and pigs [111]. Similar to other organs, diaphragm also has its own niche. Congenital defects in diaphragm can affect diaphragm functions. In the present scenario functional restoration of congenital diaphragm defects by surgical repair has risk of reoccurrence of defects or incomplete restoration [8]. Decellularization of donor derived diaphragm offers a way for reconstruction of new and functionally compatible diaphragm through niche modulation. Tissue engineering technology based decellularization of diaphragm and simultaneous perfusion of bone marrow mesenchymal stem cells (BM-MSCs) facilitates regeneration of functional scaffolds of diaphragm tissues [8]. In vivo replacement of hemidiaphragm in rats with reseeded scaffolds possesses similar myography and spirometry as it has in vivo in donor rats. These scaffolds retaining natural architecture are devoid of immune cells, retaining intact extracellular matrix that supports adhesion, proliferation, and differentiation of seeded cells [8]. These findings suggest that cadaver obtained diaphragm, seeded with BM-MSCs, can be used for curing patients in need for restoration of diaphragm functions (; ). However, BMSCs are heterogeneous population, which might result in differential outcomes in clinical settings; however clonal expansion of BMSCs yields homogenous cells population for therapeutic application [8]. One study also finds that intracavernous delivery of single clone BMSCs can restore erectile function in diabetic mice [112] and the same strategy might be explored for adult human individuals. The infection of hepatitis C virus (HCV) can cause liver cirrhosis and degeneration of hepatic tissue. The intraparenchymal transplantation of bone marrow mononuclear cells (BMMNCs) into liver tissue decreases aspartate aminotransferase (AST), alanine transaminase (ALT), bilirubin, CD34, and -SMA, suggesting that transplanted BMSCs restore hepatic functions through regeneration of hepatic tissues [113]. In order to meet the growing demand for stem cells transplantation therapy, donor encouragement is always required [8]. The stem cells donation procedure is very simple; with consent donor gets an injection of granulocyte-colony stimulating factor (G-CSF) that increases BMSCs population. Bone marrow collection is done from hip bone using syringe in 4-5hrs, requiring local anaesthesia and within a wk time frame donor gets recovered donation associated weakness.

The field of iPSCs technology and research is new to all other stem cells research, emerging in 2006 when, for the first time, Takahashi and Yamanaka generated ESCs-like cells through genetic incorporation of four factors, Sox2, Oct3/4, Klf4, and c-Myc, into skin fibroblast [3]. Due to extensive nuclear reprogramming, generated iPSCs are indistinguishable from ESCs, for their transcriptome profiling, epigenetic markings, and functional competence [3], but use of retrovirus in transdifferentiation approach has questioned iPSCs technology. Technological advancement has enabled generation of iPSCs from various kinds of adult cells phasing through ESCs or direct transdifferentiation. This section of review outlines most recent advancement in iPSC technology and regenerative applications (; ). Using the new edge of iPSCs technology, terminally differentiated skin cells directly can be transformed into kidney organoids [114], which are functionally and structurally similar to those of kidney tissue in vivo. Up to certain extent kidneys heal themselves; however natural regeneration potential cannot meet healing for severe injuries. During kidneys healing process, a progenitor stem cell needs to become 20 types of cells, required for waste excretion, pH regulation, and restoration of water and electrolytic ions. The procedure for generation of kidney organoids ex vivo, containing functional nephrons, has been identified for human. These ex vivo kidney organoids are similar to fetal first-trimester kidneys for their structure and physiology. Such kidney organoids can serve as model for nephrotoxicity screening of drugs, disease modelling, and organ transplantation. However generation of fully functional kidneys is a far seen event with today's scientific technologies [114]. Loss of neurons in age-related macular degeneration (ARMD) is the common cause of blindness. At preclinical level, transplantation of iPSCs derived neuronal progenitor cells (NPCs) in rat limits progression of disease through generation of 5-6 layers of photoreceptor nuclei, restoring visual acuity [78]. The various approaches of iPSCs mediated retinal regeneration including ARMD have been reviewed elsewhere [79]. Placenta, the cordial connection between mother and developing fetus, gets degenerated in certain pathophysiological conditions. Nuclear programming of OCT4 knock-out (KO) and wild type (WT) mice fibroblast through transient expression of GATA3, EOMES, TFAP2C, and +/ cMYC generates transgene independent trophoblast stem-like cells (iTSCs), which are highly similar to blastocyst derived TSCs for DNA methylation, H3K7ac, nucleosome deposition of H2A.X, and other epigenetic markings. Chimeric differentiation of iTSCs specifically gives rise to haemorrhagic lineages and placental tissue, bypassing pluripotency phase, opening an avenue for generation of fully functional placenta for human [115]. Neurodegenerative disease like Alzheimer's and obstinate epilepsies can degenerate cerebrum, controlling excitatory and inhibitory signals of the brain. The inhibitory tones in cerebral cortex and hippocampus are accounted by -amino butyric acid secreting (GABAergic) interneurons (INs). Loss of these neurons often leads to progressive neurodegeneration. Genomic integration of Ascl1, Dlx5, Foxg1, and Lhx6 to mice and human fibroblast transforms these adult cells into GABAergic-INs (iGABA-INs). These cells have molecular signature of telencephalic INs, release GABA, and show inhibition to host granule neuronal activity [81]. Transplantation of these INs in developing embryo cures from genetic and acquired seizures, where transplanted cells disperse and mature into functional neuronal circuits as local INs [82]. Dorsomorphin and SB-431542 mediated inhibition of TGF- and BMP signalling direct transformation of human iPSCs into cortical spheroids. These cortical spheroids consisted of both peripheral and cortical neurons, surrounded by astrocytes, displaying transcription profiling and electrophysiology similarity with developing fetal brain and mature neurons, respectively [83]. The underlying complex biology and lack of clear etiology and genetic reprogramming and difficulty in recapitulation of brain development have barred understanding of pathophysiology of autism spectrum disorder (ASD) and schizophrenia. 3D organoid cultures of ASD patient derived iPSC generate miniature brain organoid, resembling fetal brain few months after gestation. The idiopathic conditions of these organoids are similar with brain of ASD patients; both possess higher inhibitory GABAergic neurons with imbalanced neuronal connection. Furthermore these organoids express forkhead Box G1 (FOXG1) much higher than normal brain tissue, which explains that FOXG1 might be the leading cause of ASD [84]. Degeneration of other organs and tissues also has been reported, like degeneration of lungs which might occur due to tuberculosis infection, fibrosis, and cancer. The underlying etiology for lung degeneration can be explained through organoid culture. Coaxing of iPSC into inert biomaterial and defined culture leads to formation of lung organoids that consisted of epithelial and mesenchymal cells, which can survive in culture for months. These organoids are miniature lung, resemble tissues of large airways and alveoli, and can be used for lung developmental studies and screening of antituberculosis and anticancer drugs [87]. The conventional multistep reprogramming for iPSCs consumes months of time, while CRISPER-Cas9 system based episomal reprogramming system that combines two steps together enables generation of ESCs-like cells in less than twowks, reducing the chances of culture associated genetic abrasions and unwanted epigenetic [80]. This approach can yield single step ESCs-like cells in more personalized way from adults with retinal degradation and infants with severe immunodeficiency, involving correction for genetic mutation of OCT4 and DNMT3B [80]. The iPSCs expressing anti-CCR5-RNA, which can be differentiated into HIV1 resistant macrophages, have applications in AIDS therapeutics [88]. The diversified immunotherapeutic application of iPSCs has been reviewed elsewhere [89]. The -1 antitrypsin deficiency (A1AD) encoded by serpin peptidase inhibitor clade A member 1 (SERPINA1) protein synthesized in liver protects lungs from neutrophils elastase, the enzyme causing disruption of lungs connective tissue. A1AD deficiency is common cause of both lung and liver disease like chronic obstructive pulmonary disease (COPD) and liver cirrhosis. Patient specific iPSCs from lung and liver cells might explain pathophysiology of A1AD deficiency. COPD patient derived iPSCs show sensitivity to toxic drugs which explains that actual patient might be sensitive in similar fashion. It is known that A1AD deficiency is caused by single base pair mutation and correction of this mutation fixes the A1AD deficiency in hepatic-iPSCs [85]. The high order brain functions, like emotions, anxiety, sleep, depression, appetite, breathing heartbeats, and so forth, are regulated by serotonin neurons. Generation of serotonin neurons occurs prior to birth, which are postmitotic in their nature. Any sort of developmental defect and degeneration of serotonin neurons might lead to neuronal disorders like bipolar disorder, depression, and schizophrenia-like psychiatric conditions. Manipulation of Wnt signalling in human iPSCs in defined culture conditions leads to an in vitro differentiation of iPSCs to serotonin-like neurons. These iPSCs-neurons primarily localize to rhombomere 2-3 segment of rostral raphe nucleus, exhibit electrophysiological properties similar to serotonin neurons, express hydroxylase 2, the developmental marker, and release serotonin in dose and time dependent manner. Transplantation of these neurons might cure from schizophrenia, bipolar disorder, and other neuropathological conditions [116]. The iPSCs technology mediated somatic cell reprogramming of ventricular monocytes results in generation of cells, similar in morphology and functionality with PCs. SA note transplantation of PCs to large animals improves rhythmic heart functions. Pacemaker needs very reliable and robust performance so understanding of transformation process and site of transplantation are the critical aspect for therapeutic validation of iPSCs derived PCs [28]. Diabetes is a major health concern in modern world, and generation of -cells from adult tissue is challenging. Direct reprogramming of skin cells into pancreatic cells, bypassing pluripotency phase, can yield clinical grade -cells. This reprogramming strategy involves transformation of skin cells into definitive endodermal progenitors (cDE) and foregut like progenitor cells (cPF) intermediates and subsequent in vitro expansion of these intermediates to become pancreatic -cells (cPB). The first step is chemically complex and can be understood as nonepisomal reprogramming on day one with pluripotency factors (OCT4, SOX2, KLF4, and hair pin RNA against p53), then supplementation with GFs and chemical supplements on day seven (EGF, bFGF, CHIR, NECA, NaB, Par, and RG), and two weeks later (Activin-A, CHIR, NECA, NaB, and RG) yielding DE and cPF [86]. Transplantation of cPB yields into glucose stimulated secretion of insulin in diabetic mice defines that such cells can be explored for treatment of T1DM and T2DM in more personalized manner [86]. iPSCs represent underrated opportunities for drug industries and clinical research laboratories for development of therapeutics, but safety concerns might limit transplantation applications (; ) [117]. Transplantation of human iPSCs into mice gastrula leads to colonization and differentiation of cells into three germ layers, evidenced with clinical developmental fat measurements. The acceptance of human iPSCs by mice gastrula suggests that correct timing and appropriate reprogramming regime might delimit human mice species barrier. Using this fact of species barrier, generation of human organs in closely associated primates might be possible, which can be used for treatment of genetic factors governed disease at embryo level itself [118]. In summary, iPSCs are safe and effective for treatment of regenerative medicine.

The unstable growth of human population threatens the existence of wildlife, through overexploitation of natural habitats and illegal killing of wild animals, leading many species to face the fate of being endangered and go for extinction. For wildlife conservation, the concept of creation of frozen zoo involves preservation of gene pool and germ plasm from threatened and endangered species (). The frozen zoo tissue samples collection from dead or live animal can be DNA, sperms, eggs, embryos, gonads, skin, or any other tissue of the body [119]. Preserved tissue can be reprogrammed or transdifferentiated to become other types of tissues and cells, which opens an avenue for conservation of endangered species and resurrection of life (). The gonadal tissue from young individuals harbouring immature tissue can be matured in vivo and ex vivo for generation of functional gametes. Transplantation of SSCs to testis of male from the same different species can give rise to spermatozoa of donor cells [120], which might be used for IVF based captive breeding of wild animals. The most dangerous fact in wildlife conservation is low genetic diversity, too few reproductively capable animals which cannot maintain adequate genetic diversity in wild or captivity. Using the edge of iPSC technology, pluripotent stem cells can be generated from skin cells. For endangered drill, Mandrillus leucophaeus, and nearly extinct white rhinoceros, Ceratotherium simum cottoni, iPSC has been generated in 2011 [121]. The endangered animal drill (Mandrillus leucophaeus) is genetically very close to human and often suffers from diabetes, while rhinos are genetically far removed from other primates. The progress in iPSCs, from the human point of view, might be transformed for animal research for recapturing reproductive potential and health in wild animals. However, stem cells based interventions in wild animals are much more complex than classical conservation planning and biomedical research has to face. Conversion of iPSC into egg or sperm can open the door for generation of IVF based embryo; those might be transplanted in womb of live counterparts for propagation of population. Recently, iPSCs have been generated for snow leopard (Panthera uncia), native to mountain ranges of central Asia, which belongs to cat family; this breakthrough has raised the possibilities for cryopreservation of genetic material for future cloning and other assisted reproductive technology (ART) applications, for the conservation of cat species and biodiversity. Generation of leopard iPSCs has been achieved through retroviral-system based genomic integration of OCT4, SOX2, KLF4, cMYC, and NANOG. These iPSCs from snow leopard also open an avenue for further transformation of iPSCs into gametes [122]. The in vivo maturation of grafted tissue depends both on age and on hormonal status of donor tissue. These facts are equally applicable to accepting host. Ectopic xenografts of cryopreserved testis tissue from Indian spotted deer (Moschiola indica) to nude mice yielded generation of spermatocytes [123], suggesting that one-day procurement of functional sperm from premature tissue might become a general technique in wildlife conservation. In summary, tissue biopsies from dead or live animals can be used for generation of iPSCs and functional gametes; those can be used in assisted reproductive technology (ART) for wildlife conservation.

The spectacular progress in the field of stem cells research represents great scope of stem cells regenerative therapeutics. It can be estimated that by 2020 or so we will be able to produce wide array of tissue, organoid, and organs from adult stem cells. Inductions of pluripotency phenotypes in terminally differentiated adult cells have better therapeutic future than ESCs, due to least ethical constraints with adult cells. In the coming future, there might be new pharmaceutical compounds; those can activate tissue specific stem cells, promote stem cells to migrate to the side of tissue injury, and promote their differentiation to tissue specific cells. Except few countries, the ongoing financial and ethical hindrance on ESCs application in regenerative medicine have more chance for funding agencies to distribute funding for the least risky projects on UCSCs, BMSCs, and TSPSCs from biopsies. The existing stem cells therapeutics advancements are more experimental and high in cost; due to that application on broad scale is not feasible in current scenario. In the near future, the advancements of medical science presume using stem cells to treat cancer, muscles damage, autoimmune disease, and spinal cord injuries among a number of impairments and diseases. It is expected that stem cells therapies will bring considerable benefits to the patients suffering from wide range of injuries and disease. There is high optimism for use of BMSCs, TSPSCs, and iPSCs for treatment of various diseases to overcome the contradictions associated with ESCs. For advancement of translational application of stem cells, there is a need of clinical trials, which needs funding rejoinder from both public and private organizations. The critical evaluation of regulatory guidelines at each phase of clinical trial is a must to comprehend the success and efficacy in time frame.

Dr. Anuradha Reddy from Centre for Cellular and Molecular Biology Hyderabad and Mrs. Sarita Kumari from Department of Yoga Science, BU, Bhopal, India, are acknowledged for their critical suggestions and comments on paper.

There are no competing interests associated with this paper.

Visit link:
Stem Cells Applications in Regenerative Medicine and ...

Read More...

Regenerative & Sports Medicine | Dr. Rand McClain

Sunday, November 7th, 2021

label, .bookly-form .bookly-label-error, .bookly-form .bookly-progress-tracker > .active, .bookly-form .picker__nav--next, .bookly-form .pickadate__nav--prev, .bookly-form .picker__day:hover, .bookly-form .picker__day--selected:hover, .bookly-form .picker--opened .picker__day--selected, .bookly-form .picker__button--clear, .bookly-form .picker__button--today, .bookly-form .bookly-columnizer .bookly-hour span.bookly-waiting-list { color: #f4662f!important; } /* Background */ .bookly-form .bookly-progress-tracker > .active .step, .bookly-form .picker__frame, .bookly-form .bookly-service-step .bookly-week-days label, .bookly-form .bookly-repeat-step .bookly-week-days label, .bookly-form .bookly-columnizer .bookly-hour:hover .bookly-hour-icon span, .bookly-form .bookly-btn, .bookly-form .bookly-btn:active, .bookly-form .bookly-btn:focus, .bookly-form .bookly-btn:hover, .bookly-form .bookly-btn-submit, .bookly-form .bookly-round, .bookly-form .bookly-square { background-color: #f4662f!important; } .bookly-form .bookly-triangle { border-bottom-color: #f4662f!important; } /* Border */ .bookly-form input[type="text"].bookly-error, .bookly-form input[type="password"].bookly-error, .bookly-form select.bookly-error, .bookly-form textarea.bookly-error, .bookly-extra-step div.bookly-extras-thumb.bookly-extras-selected { border: 2px solid #f4662f!important; } /* Other */ .bookly-form .picker__header { border-bottom: 1px solid #f4662f!important; } .bookly-form .picker__nav--next:before { border-left: 6px solid #f4662f!important; } .bookly-form .picker__nav--prev:before { border-right: 6px solid #f4662f!important; } .bookly-form .bookly-service-step .bookly-week-days label.active,.bookly-form .bookly-repeat-step .bookly-week-days label.active { background: #f4662f url(https://psrmed.com/wp-content/plugins/bookly-responsive-appointment-booking-tool/frontend/resources/images/checkbox.png) 0 0 no-repeat!important; } .bookly-form .bookly-pagination > li.active,.bookly-form .bookly-columnizer .bookly-day,.bookly-form .bookly-schedule-date { background: #f4662f!important; border: 1px solid #f4662f!important; } .bookly-form .bookly-columnizer .bookly-hour:hover { border: 2px solid #f4662f!important; color: #f4662f!important; } .bookly-form .bookly-columnizer .bookly-hour:hover .bookly-hour-icon { background: none; border: 2px solid #f4662f!important; color: #f4662f!important; }

View post:
Regenerative & Sports Medicine | Dr. Rand McClain

Read More...

AMSSM Releases Position Statement on Regenerative Medicine in Sports Medicine – Newswise

Sunday, November 7th, 2021

Newswise The American Medical Society for Sports Medicine (AMSSM) has released a position statement on Principles for the Responsible Use of Regenerative Medicine in Sports Medicine.

This position statement provides sports medicine physicians with information on regenerative medicine terminology, a brief review of the basic science and clinical studies, regulatory considerations, and best practices for introducing the orthobiologic classification of regenerative therapies into their clinical practice.

The document is being published in the Clinical Journal of Sport Medicine, with accompanying editorial highlights published in the British Journal of Sports Medicine. Both are freely accessible on their respective websites.

Sports medicine physicians would benefit from decision-making guidance about whether to introduce orthobiologics into their practice and how to do it responsibly, said Dr. Jonathan Finnoff, the Chief Medical Officer of the United States Olympic and Paralympic Committee and the lead author of the statement. The information within this statement will help sports medicine physicians make informed and responsible decisions about the role of regenerative medicine and orthobiologics in their practice.

In 2019, the AMSSM Board of Directors established a Regenerative Medicine Task Force, with a subgroup charged to develop a regenerative medicine position statement. The Task Force brought together a writing group that included sports medicine physicians and scientists who are recognized leaders in bioethics, research, and regenerative medicine clinical applications to produce this statement.

The field of regenerative medicine, and the sub-classification of orthobiologics, involves a variety of therapies and techniques focused on the repair or replacement of damaged or diseased tissue to restore function. Despite these novel therapies being very attractive to sports medicine physicians and patients alike, this is a complex and controversial topic.

Common orthobiologics that are employed in research and medical practice are being combined under the umbrella of stem cell therapy in a manner that is confusing to both patients and the public, said Dr. Shane Shapiro, one of the lead authors of the statement. The need for scientifically validated treatments for non-healing orthopedic and sports conditions has increased interest in orthobiologics and other regenerative therapies to address existing treatment gaps.

The document contains brief discussions of the basic science, proposed therapeutic mechanisms of action, and clinical evidence related to regenerative medicine products, including uses for platelet-rich plasma and other cellular therapies. Additionally, the statement features sections regarding regulatory considerations and an in-depth portion on introducing regenerative medicine into clinical practice.

Ultimately, this AMSSM position statement on regenerative medicine advocates for the advancement of orthobiologic science, patient safety and education towards the responsible translation of regenerative therapies, said Dr. Kenneth Mautner, co-lead author of the position statement and an AMSSM Board member.

About the AMSSM: AMSSM is a multi-disciplinary organization of sports medicine physicians dedicated to education, research, advocacy and the care of athletes of all ages. The majority of AMSSM members are primary care physicians with fellowship training and added qualification in sports medicine who then combine their practice of sports medicine with their primary specialty. AMSSM includes members who specialize solely in non-surgical sports medicine and serve as team physicians at the youth level, NCAA, NFL, MLB, NBA, WNBA, MLS and NHL, as well as with Olympic and Paralympic teams. By nature of their training and experience, sports medicine physicians are ideally suited to provide comprehensive medical care for athletes, sports teams or active individuals who are simply looking to maintain a healthy lifestyle. http://www.amssm.org

Read the original here:
AMSSM Releases Position Statement on Regenerative Medicine in Sports Medicine - Newswise

Read More...

CRISPR Therapeutics Provides Business Update and Reports Third Quarter 2021 Financial Results – Yahoo Finance

Sunday, November 7th, 2021

-Achieved target enrollment in CTX001 clinical trials for beta thalassemia (TDT) and sickle cell disease (SCD); regulatory submissions planned for late 2022-

-Reported positive results from the ongoing Phase 1 CARBON clinical trial evaluating the safety and efficacy of CTX110 for CD19+ B-cell malignancies; enrollment continues, with potential registrational trial incorporating consolidation dosing expected to initiate in Q1 2022-

-Implementing consolidation dosing protocols for CTX120 and CTX130 clinical trials; enrollment continues, with top-line data expected to report in 1H 2022-

-Regenerative medicine and in vivo programs continue to progress and remain on track-

ZUG, Switzerland and CAMBRIDGE, Mass., Nov. 03, 2021 (GLOBE NEWSWIRE) -- CRISPR Therapeutics (Nasdaq: CRSP), a biopharmaceutical company focused on creating transformative gene-based medicines for serious diseases, today reported financial results for the third quarter ended September 30, 2021.

The third quarter marked significant progress across our portfolio, said Samarth Kulkarni, Ph.D., Chief Executive Officer of CRISPR Therapeutics. With our partner Vertex, we achieved target enrollment for the CTX001 clinical trials in patients with beta thalassemia and sickle cell disease, which can support regulatory submissions in late 2022. Additionally, we demonstrated proof of concept for our allogeneic CAR-T platform with positive data from our CARBON trial of CTX110, which showed that immediately available off-the-shelf cell therapies can offer efficacy similar to autologous CAR-T with a differentiated safety profile for patients with large B-cell lymphomas. Based on these encouraging results, we plan to expand the CARBON trial into a potentially registrational trial in the first quarter of 2022. Furthermore, we hope to bring these transformative allogeneic CAR-T therapies to patients in outpatient and community oncology settings, enabling broad access."

Story continues

Recent Highlights and Outlook

Third Quarter 2021 Financial Results

Cash Position: Cash, cash equivalents and marketable securities were $2,477.4 million as of September 30, 2021, compared to $2,589.4 million as of June 30, 2021. The decrease in cash of $112.0 million was primarily driven by cash used in operating activities to support ongoing research and development of the Companys clinical and pre-clinical programs.

Revenue: Total collaboration revenue was $0.3 million for the third quarter of 2021, compared to $0.1 million for the third quarter of 2020. Collaboration revenue primarily consisted of revenue recognized in connection with our collaboration agreements with Vertex.

R&D Expenses: R&D expenses were $105.3 million for the third quarter of 2021, compared to $71.0 million for the third quarter of 2020. The increase in expense was driven by development activities supporting the advancement of the hemoglobinopathies program and wholly-owned immuno-oncology programs, as well as increased headcount and supporting facilities related expenses.

G&A Expenses: General and administrative expenses were $24.4 million for the third quarter of 2021, compared to $21.5 million for the third quarter of 2020. The increase in general and administrative expenses for the year was primarily driven by headcount-related expense.

Net Loss: Net loss was $127.2 million for the third quarter of 2021, compared to a net loss of $92.4 million for the third quarter of 2020.

About CTX001CTX001 is an investigational, autologous, ex vivo CRISPR/Cas9 gene-edited therapy that is being evaluated for patients suffering from TDT or severe SCD, in which a patients hematopoietic stem cells are edited to produce high levels of fetal hemoglobin (HbF; hemoglobin F) in red blood cells. HbF is a form of the oxygen-carrying hemoglobin that is naturally present at birth, which then switches to the adult form of hemoglobin. The elevation of HbF by CTX001 has the potential to alleviate or eliminate transfusion requirements for patients with TDT and reduce or eliminate painful and debilitating sickle crises for patients with SCD. Earlier results from these ongoing trials were published as a Brief Report in The New England Journal of Medicine in January of 2021.

Based on progress in this program to date, CTX001 has been granted Regenerative Medicine Advanced Therapy (RMAT), Fast Track, Orphan Drug, and Rare Pediatric Disease designations from the U.S. Food and Drug Administration (FDA) for both TDT and SCD. CTX001 has also been granted Orphan Drug Designation from the European Commission, as well as Priority Medicines (PRIME) designation from the European Medicines Agency (EMA), for both TDT and SCD.

Among gene-editing approaches being investigated/evaluated for TDT and SCD, CTX001 is the furthest advanced in clinical development.

About the CRISPR-Vertex CollaborationVertex and CRISPR Therapeutics entered into a strategic research collaboration in 2015 focused on the use of CRISPR/Cas9 to discover and develop potential new treatments aimed at the underlying genetic causes of human disease. CTX001 represents the first potential treatment to emerge from the joint research program. Under a recently amended collaboration agreement, Vertex will lead global development, manufacturing and commercialization of CTX001 and split program costs and profits worldwide 60/40 with CRISPR Therapeutics.

About CLIMB-111The ongoing Phase 1/2 open-label trial, CLIMB-Thal-111, is designed to assess the safety and efficacy of a single dose of CTX001 in patients ages 12 to 35 with TDT. The trial will enroll up to 45 patients and follow patients for approximately two years after infusion. Each patient will be asked to participate in a long-term follow-up trial.

About CLIMB-121The ongoing Phase 1/2 open-label trial, CLIMB-SCD-121, is designed to assess the safety and efficacy of a single dose of CTX001 in patients ages 12 to 35 with severe SCD. The trial will enroll up to 45 patients and follow patients for approximately two years after infusion. Each patient will be asked to participate in a long-term follow-up trial.

About CLIMB-131This is a long-term, open-label trial to evaluate the safety and efficacy of CTX001 in patients who received CTX001 in CLIMB-111 or CLIMB-121. The trial is designed to follow participants for up to 15 years after CTX001 infusion.

About CTX110CTX110, a wholly owned program of CRISPR Therapeutics, is a healthy donor-derived gene-edited allogeneic CAR-T investigational therapy targeting cluster of differentiation 19, or CD19. CTX110 is being investigated in the ongoing CARBON trial.

About CARBONThe ongoing Phase 1 single-arm, multi-center, open label clinical trial, CARBON, is designed to assess the safety and efficacy of several dose levels of CTX110 for the treatment of relapsed or refractory B-cell malignancies.

About CTX120CTX120, a wholly-owned program of CRISPR Therapeutics, is a healthy donor-derived gene-edited allogeneic CAR-T investigational therapy targeting B-cell maturation antigen, or BCMA. CTX120 is being investigated in an ongoing Phase 1 single-arm, multi-center, open-label clinical trial designed to assess the safety and efficacy of several dose levels of CTX120 for the treatment of relapsed or refractory multiple myeloma. CTX120 has been granted Orphan Drug designation from the FDA.

About CTX130CTX130, a wholly-owned program of CRISPR Therapeutics, is a healthy donor-derived gene-edited allogeneic CAR-T investigational therapy targeting cluster of differentiation 70, or CD70, an antigen expressed on various solid tumors and hematologic malignancies. CTX130 is being developed for the treatment of both solid tumors, such as renal cell carcinoma, and T-cell and B-cell hematologic malignancies. CTX130 is being investigated in two ongoing independent Phase 1, single-arm, multi-center, open-label clinical trials that are designed to assess the safety and efficacy of several dose levels of CTX130 for the treatment of relapsed or refractory renal cell carcinoma and various subtypes of lymphoma, respectively.

About CRISPR TherapeuticsCRISPR 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.

CRISPR THERAPEUTICS word mark and design logo, CTX001, CTX110, CTX120, and CTX130 are trademarks and registered trademarks of CRISPR Therapeutics AG. All other trademarks and registered trademarks are the property of their respective owners.

CRISPR Therapeutics Forward-Looking StatementThis 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 in this press release, as well as statements regarding CRISPR Therapeutics expectations about any or all of the following: (i) the safety, efficacy, data and clinical progress of CRISPR Therapeutics various clinical programs, including CTX001, CTX110, CTX120 and CTX130; (ii) the status of clinical trials and preclinical studies (including, without limitation, the expected timing of data releases and development, as well as initiation and completion of clinical trials) and development timelines for CRISPR Therapeutics product candidates; (iii) expectations regarding the data that has been presented from our various clinical trials (including our CARBON trial) as well as 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 or to support regulatory filings; (iv) the actual or potential benefits of regulatory designations; (v) the potential benefits of CRISPR Therapeutics collaborations and strategic partnerships; (vi) the intellectual property coverage and positions of CRISPR Therapeutics, its licensors and third parties as well as the status and potential outcome of proceedings involving any such intellectual property; (vii) the sufficiency of CRISPR Therapeutics cash resources; and (viii) 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; that one or more of CRISPR Therapeutics internal or external product candidate programs will not proceed as planned for technical, scientific or commercial reasons; that future competitive or other market factors may adversely affect the commercial potential for CRISPR Therapeutics product candidates; uncertainties inherent in the initiation and completion of preclinical studies for CRISPR Therapeutics product candidates (including, without limitation, availability and timing of results and whether such results will be predictive of future results of the future trials); uncertainties about regulatory approvals to conduct trials or to market products; the potential impacts due to the coronavirus pandemic such as (x) delays in regulatory review, manufacturing and supply chain interruptions, adverse effects on healthcare systems and disruption of the global economy; (y) the timing and progress of clinical trials, preclinical studies and other research and development activities; and (z) the overall impact of the coronavirus pandemic on its business, financial condition and results of operations; 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.

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

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

CRISPR Therapeutics AGCondensed Consolidated Statements of Operations(Unaudited, In thousands except share data and per share data)

Three Months Ended September 30,

Nine Months Ended September 30,

2021

2020

2021

2020

Revenue:

Collaboration revenue

$

329

$

148

$

900,733

$

349

Grant revenue

495

1,331

Total revenue

$

824

$

148

$

902,064

$

349

Operating expenses:

Research and development

105,321

71,008

304,163

184,581

General and administrative

24,352

21,539

78,675

62,442

Total operating expenses

129,673

92,547

382,838

247,023

(Loss) income from operations

(128,849

)

(92,399

)

519,226

(246,674

)

Total other income, net

1,101

160

3,806

5,804

Net (loss) income before income taxes

(127,748

)

(92,239

)

523,032

(240,870

)

Benefit (provision) for income taxes

595

Continued here:
CRISPR Therapeutics Provides Business Update and Reports Third Quarter 2021 Financial Results - Yahoo Finance

Read More...

ORGANICELL REGENERATIVE MEDICINE, INC. : Entry into a Material Definitive Agreement, Unregistered Sale of Equity Securities, Financial Statements and…

Sunday, November 7th, 2021

Item 1.01 Entry into a Material Definitive Agreement.

On October 29, 2021, the Company entered into an Exchange Agreement (the"Exchange Agreement") with shareholders who were issued shares under (i) variousconsulting and employment agreements during 2021 (the "Service Providers"), and(ii) those shareholders who were issued shares of common stock pursuant to theCompany's Management and Consultants Performance Stock Plan (the "MCP Plan")(each person who received shares pursuant to the MCP Plan is referred to as an"MCP Plan Holder").

The Service Providers who executed the Exchange Agreement were issued a total of30,300,000 shares under their respective consulting or employment agreements(the "Service Provider Shares"), and the MCP Plan Holders who executed theExchange Agreement received a total of 49,500,000 shares under the MCP Plan, foran aggregate of 79,800,000 shares of common stock. As of the effective date ofthe Agreement, the Service Providers and MCP Plan Holders who executed theExchange Agreement agreed to exchange their respective Service Provider Sharesor the shares issued under the MCP Plan for newly issued shares pursuant to theCompany's newly formed 2021 Equity Incentive Plan (the "EIP"), on a 1:1 basis,resulting in the issuance of 79,800,000 shares of common stock under the EIP(the "Exchange Shares").

The Exchange Agreement contains certain customary representations, warranties,and covenants for transactions of this type.

The description of the Exchange Agreement does not purport to be complete and isqualified in its entirety by reference to the full text of the form of ExchangeAgreement which is attached as Exhibit 10.1 to this Current Report on Form8-K and is incorporated herein by reference.

Item 3.02 Unregistered Sales of Equity Securities.

The disclosure set forth above in Item 1.01 of this Current Report on Form 8-Kwith respect to the issuances of the Exchange Sharers pursuant to the ExchangeAgreement is incorporated by reference into this Item 3.02.

The Exchange Shares were issued in reliance on the exemption from registrationrequirements thereof provided by Section 4(a)(2) of the Securities Act.

Item 9.01 Financial Statements and Exhibits.

* Schedules, exhibits and similar attachments have been omitted pursuant to Item601(a)(5) of Regulation S-K. The Company hereby undertakes to furnish copies ofsuch omitted materials supplementally upon request by the U.S. Securities andExchange Commission.

1

Edgar Online, source Glimpses

See original here:
ORGANICELL REGENERATIVE MEDICINE, INC. : Entry into a Material Definitive Agreement, Unregistered Sale of Equity Securities, Financial Statements and...

Read More...

Blood Thawing System Market to Witness Massive Revenue Streams from Growing Demand for Rapid Dry Plasma Thawing Products for FFPs in Blood Banks,…

Sunday, November 7th, 2021

ALBANY, N.Y., Nov. 3, 2021 /PRNewswire/ -- Advancements in blood thawing devices have pivoted on the changing requirements of fresh frozen plasma (FFP) in transfusion practices and thawing procedures in cryopreservation. Plasma thawers to maintain the integrity of FFPs are growing in applications in laboratories, blood banks, and hospital settings. New procedures and technologies have been introduced in the blood thawing system market, which prevent potential risks of contamination. Particularly, the adoption of dry bathing systems for preventing transfusion-associated bacterial sepsis in treating blood disorders and cancer is gaining momentum.

The use of FDA-approved, CE-marked, and ISO certified plasma thawers are gaining popularity in umbilical cord blood processing and cell-based therapies, thereby enriching the prospects of regenerative medicine. The need for new device designs and software for temperature controllers occupies a key role in improving the existing cryopreservation protocols, which has opened up a lucrative avenue for players, notes the study on the blood thawing systems market.

Request Brochure of Blood Thawing System Market Research Report - https://www.transparencymarketresearch.com/sample/sample.php?flag=B&rep_id=81455

Dry plasma thawing products are gaining preference over traditional water baths or wet plasma thawers, as they help increase the success of transfusion of adult stem cells. Asia Pacific is a highly lucrative market, where the players have gained opportunities from rising on-demand cell thawing to successfully deliver cell-based therapies to target population. The global valuation of the blood thawing system market is projected to reach US$ 400 Mn by 2030, at a CAGR of 7.7% during the forecast period.

Key Findings of Blood Thawing System Market Study

GMP-compliant Equipment Improve Safety and Effectiveness of Thawing Processes: End users have become increasingly aware about Good Manufacturing Practice (GMP), manufacturer's instructions, and other guidelines for preparing FFPs for use in various applications of transfusion medicine. Most prominently, the risk of transfusion-associated bacterial sepsis has led to constant technological advancements in the blood thawing equipment to ensure the efficacy of the thawing method. GMP-ready cryochain hardware and software are gaining traction in umbilical cord blood processing, finds a TMR study on the blood thawing system market.

Request for Analysis of COVID-19 Impact on Blood Thawing System Market https://www.transparencymarketresearch.com/sample/sample.php?flag=covid19&rep_id=81455

The success of cryopreservation is dependent on appropriate execution of thawing procedures. Indeed, advances made in the protocols for on-demand thawing in regenerative medicine have bolstered the prospects of the blood thawing system market.

Regulatory Approval of Next-generation Thawing Devices Extends Horizon: The need for eliminating continuous blood products manufacturing is a key underpinning for adopting reliable thawing processes. In this light, thawing equipment for rapid, reliable thawing of FFP is garnering attention of clinicians for use in patients in emergency setting as well as for meeting the demand in planned hospitalizations.

Next-gen plasma thawing devices promise low turnaround time, are of portable designs, and ensure high throughput. The use of such devices is expected to rise in various applications in exchange transfusions, stem cell transfusions, and crystalloid infusion solutions.

TMR offers custom market research services that help clients to get information on their business scenario required where syndicated solutions are not enough, Request for Custom Research- https://www.transparencymarketresearch.com/sample/sample.php?flag=CR&rep_id=81455

Blood Thawing System Market: Key Drivers

Buy Blood Thawing System Market Report https://www.transparencymarketresearch.com/checkout.php?rep_id=81455&ltype=S

Blood Thawing System Market: Regional Dynamics

Blood Thawing System Market: Key Players

Some of the key players in the blood thawing system market are Sartorius AG, Thermo Fisher Scientific, Inc., Cytiva (GE Healthcare), Cardinal Health, KW Scientific Apparatus Srl, Boekel Scientific, Barkey GmbH & Co. KG, Helmer Scientific Inc., and Fremon Scientific Inc.

Global Blood Thawing System Market: Segmentation

Modernization of healthcare in terms of both infrastructure and services have pushed the healthcare industry to new heights, Stay Updated with Latest Healthcare Industry Research Reportsby Transparency Market Research:

Blood Purification Equipment Market: Increasing knowledge and understanding of pathophysiology and hematology due to additional research and development coupled with noteworthy progress in bio separation techniques are some of the other factors contributing to the overall growth of the global blood purification equipment market

Autoimmune Disease Diagnostics Market: Increasing awareness and knowledge about autoimmune diseases among patients and care-givers would significantly contribute to the growth of the autoimmune disease diagnostics market. Rising awareness among people and increasing government initiatives are the major factors driving the autoimmune disease diagnostics market

Capillary and Venous Blood Sampling Devices Market: Manufacturers in the capillary and venous blood sampling devices market are increasing their focus to develop COVID-19 rapid test kits that are suitable for qualitative detection of the novel coronavirus using finger-prick samples. Companies are increasing efforts to innovate in small volume blood collection devices that are being made available for retail pharmacies.

About Us

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

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

For More Research Insights on Leading Industries, Visit Our YouTube Channel and hit subscribe for Future Update -https://www.youtube.com/channel/UC8e-z-g23-TdDMuODiL8BKQ

Contact

Mr. Rohit BhiseyTransparency Market ResearchState Tower,90 State Street, Suite 700,Albany NY 12207United StatesUSA - Canada Toll Free: 866-552-3453Email: [emailprotected]Follow Us: Twitter | LinkedInBlog: https://tmrblog.comBrowse PR - https://www.transparencymarketresearch.com/pressrelease/blood-thawing-system-market.htm

SOURCE Transparency Market Research

Link:
Blood Thawing System Market to Witness Massive Revenue Streams from Growing Demand for Rapid Dry Plasma Thawing Products for FFPs in Blood Banks,...

Read More...

Page 7«..6789..2030..»


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