Page 674«..1020..673674675676..680690..»

Sio Gene Therapies (SIOX) falls 3.94% for August 16 – Equities.com

Posted: August 18, 2021 at 2:01 am

Last Price$ Last TradeChange$ Change Percent %Open$ Prev Close$ High$ low$ 52 Week High$ 52 Week Low$ Market CapPE RatioVolumeExchange

SIOX - Market Data & News

Sio Gene Therapies Inc (NASDAQ: SIOX), a 11 12 St Jamess Square, London, company, fell to close at $1.95 Monday after losing $0.08 (3.94%) on volume of 431,354 shares. The stock ranged from a high of $2.02 to a low of $1.94 while Sio Gene Therapiess market cap now stands at $142,235,939.

Sio Gene Therapies combines cutting-edge science with bold imagination to develop genetic medicines that aim to radically improve the lives of patients. Its current pipeline of clinical-stage candidates includes the first potentially curative AAV-based gene therapies for GM1 gangliosidosis and Tay-Sachs/Sandhoff diseases, which are rare and uniformly fatal pediatric conditions caused by single gene deficiencies. The company is also expanding the reach of gene therapy to highly prevalent conditions such as Parkinson's disease, which affects millions of patients globally. Led by an experienced team of gene therapy development experts, and supported by collaborations with premier academic, industry and patient advocacy organizations, Sio is focused on accelerating its candidates through clinical trials to liberate patients with debilitating diseases through the transformational power of gene therapies.

Visit Sio Gene Therapies Incs profile for more information.

The Nasdaq Stock Market is a global leader in trading data and services, and equities and options listing. Nasdaq is the world's leading exchange for options volume and is home to the five largest US companies - Apple, Microsoft, Amazon, Alphabet and Facebook.

To get more information on Sio Gene Therapies Inc and to follow the companys latest updates, you can visit the companys profile page here: Sio Gene Therapies Incs Profile. For more news on the financial markets be sure to visit Equities News. Also, dont forget to sign-up for the Daily Fix to receive the best stories to your inbox 5 days a week.

Sources: Chart is provided by TradingView based on 15-minute-delayed prices. All other data is provided by IEX Cloud as of 8:05 pm ET on the day of publication.

DISCLOSURE:The views and opinions expressed in this article are those of the authors, and do not represent the views of equities.com. Readers should not consider statements made by the author as formal recommendations and should consult their financial advisor before making any investment decisions. To read our full disclosure, please go to: http://www.equities.com/disclaimer

View post:
Sio Gene Therapies (SIOX) falls 3.94% for August 16 - Equities.com

Posted in Gene therapy | Comments Off on Sio Gene Therapies (SIOX) falls 3.94% for August 16 – Equities.com

Existing Drug May Help Improve Responses to Cellular Therapies in Advanced Leukemias – pennmedicine.org

Posted: August 18, 2021 at 2:01 am

PHILADELPHIAToo many exhausted T cells left in the wake of aggressive chemotherapy regimens for patients with advanced chronic lymphocytic leukemia (CLL) make it more challenging for chimeric antigen receptor (CAR) T cell therapy to do its job. Now, a new study from researchers in the Perelman School of Medicine at the University of Pennsylvania shows how to overcome this type of resistance and reinvigorate these T cells with an experimental small molecule inhibitor.

Reporting online today in the Journal of Clinical Investigation, the team shows how the drug, known as JQ1, improved CAR T cell function by inhibiting what is known as the bromodomain and extra terminal (BET) proteins. BET, the researchers showed, can disrupt CAR expression and key acetylated histone functions in T cells in CLL.

The findings demonstrate, for the first time, this mechanism of resistance and present a much-needed target for CLL when treating patients with cellular therapies like CAR. Only a small subset of patients with advanced CLL respond to CAR T cell therapycompared to 80 percent of acute lymphocytic leukemia patients with advanced disease.

Why CAR T cells fail to fully attack cancer cells in so many CLL patients is an important question that needs to be answered in order to expand the use of these immunotherapies in CLL and other cancers, said senior author Joseph A. Fraietta, PhD, an assistant professor of Microbiology at Penn, and member of the Center for Cellular Immunotherapies. Treating these war weary T cells during the CAR T cell engineering process has the potential to boost responses, weve shown here. Its setting the stage for a very promising set of next steps that rationalize further studies, including clinical trials, to prove this approach is safe and feasible.

Using the small molecule inhibitor and the T cells and CD19 CAR T cells from multiple previously treated patients, the researchers demonstrated that the BET protein plays a role in downregulating CAR expression, and that, if blocked, can diminish CAR cell T cell exhaustion and increase the production of CAR T cells from CLL patients with poor lymphocytes.

Treatment with JQ1 also increased levels of various immunoregulatory cytokines and chemokines previously reported to be produced by CAR T cells in CLL during successful therapy. The array of native immune and CAR cells mirrored those found more typically in patients who do respond.

Given this observed reinvigoration of dysfunctional CLL patient CAR T cells by BET inhibition, the authors suggest that incorporating JQ1 into cellular engineering and expansion processes could lead to a generation of less defective and more potent final CAR T cells for patients.

To what extent the above pathways contribute to the effects of JQ1 on CAR T cells is a focus of ongoing investigations for the research group.

This work shows us that T cells can be taught new tricks, said Bruce Levine, PhD, the Barbara and Edward Netter Professor in Cancer Gene Therapy in Penns Perelman School of Medicine, and co-author on the study.That is to say that the methods of manufacturing can be adapted to improve CAR T cell function, so that what would have been exhausted or dysfunctional cells can now be reinvigorated, and potentially lead to better clinical responses in more patients than before.

This work was supported by the Bob Levis Funding Group, along with the National Institute of Allergy and Infectious Diseases (T32 AI007632), National Cancer Institute (P01 CA214278 575, R01 CA241762 U54 CA244711 576, P30 CA016520-44S3, and P30 CA016520-44S4), National Institute on Aging (U01 AG066100), the National Institute of General Medical Sciences (R01 GM118501), an Emerging Cancer Informatics Center of Excellence award from the Penn Institute for Biomedical Informatics and Abramson Cancer Center,Gabrielles Angel Foundation, an Alliance for Cancer Gene Therapy Investigator Award in Cell and Gene Therapy for Cancer, and Novartis.

Editors note: Fraietta is a co-founder of DeCART Therapeutics, Inc. and Levine is a co-founder of Tmunity Therapeutics, Inc. The University of Pennsylvania has licensed certain study-related technologies to Novartis. Penn and the inventors of these technologies receive significant financial benefits as a result of this licensing relationship with Novartis.

Penn Medicineis one of the worlds leading academic medical centers, dedicated to the related missions of medical education, biomedical research, and excellence in patient care. Penn Medicine consists of theRaymond and Ruth Perelman School of Medicine at the University of Pennsylvania (founded in 1765 as the nations first medical school) and theUniversity of Pennsylvania Health System, which together form a $8.9 billion enterprise.

The Perelman School of Medicine has been ranked among the top medical schools in the United States for more than 20 years, according toU.S. News & World Report's survey of research-oriented medical schools. The School is consistently among the nation's top recipients of funding from the National Institutes of Health, with $496 million awarded in the 2020 fiscal year.

The University of Pennsylvania Health Systems patient care facilities include: the Hospital of the University of Pennsylvania and Penn Presbyterian Medical Centerwhich are recognized as one of the nations top Honor Roll hospitals byU.S. News & World ReportChester County Hospital; Lancaster General Health; Penn Medicine Princeton Health; and Pennsylvania Hospital, the nations first hospital, founded in 1751. Additional facilities and enterprises include Good Shepherd Penn Partners, Penn Medicine at Home, Lancaster Behavioral Health Hospital, and Princeton House Behavioral Health, among others.

Penn Medicine is powered by a talented and dedicated workforce of more than 44,000 people. The organization also has alliances with top community health systems across both Southeastern Pennsylvania and Southern New Jersey, creating more options for patients no matter where they live.

Penn Medicine is committed to improving lives and health through a variety of community-based programs and activities. In fiscal year 2020, Penn Medicine provided more than $563 million to benefit our community.

Link:
Existing Drug May Help Improve Responses to Cellular Therapies in Advanced Leukemias - pennmedicine.org

Posted in Gene therapy | Comments Off on Existing Drug May Help Improve Responses to Cellular Therapies in Advanced Leukemias – pennmedicine.org

Freeline Appoints Michael J. Parini as Chief Executive Officer and Reports Second Quarter 2021 Financial Results – GlobeNewswire

Posted: August 18, 2021 at 2:01 am

On track for three programs in the clinic by year end; trial site initiation for FLT180a for Hemophilia B and FLT201 for Gaucher disease Type 1 expected by year end; program for FLT190 for Fabry disease progressing

Completed first dose cohort for FLT190 Phase 1/2 dose-finding trial and advancing to next dose level; data presented by year end

Updated durability data from FLT180a Phase1/2 dose-finding trial presented by year end

Defined Company strategic priorities to accelerate value creation; near-term efficiencies extend cash runway into first quarter of 2023

LONDON, Aug. 16, 2021 (GLOBE NEWSWIRE) -- Freeline Therapeutics Holdings plc (Nasdaq: FRLN) (the Company or Freeline), a clinical-stage biotechnology company developing transformative AAV-mediated gene therapies for patients suffering from inherited systemic debilitating diseases, today announced that Michael J. Parini has succeeded Theresa Heggie as Chief Executive Officer (CEO). The Company also reported financial results for the second quarter of 2021 and provided updates on its lead program FLT180a for Hemophilia B, FLT190 for Fabry disease, FLT201 for Gaucher disease Type 1 and FLT210 for Hemophilia A.

Today we are pleased to announce that Freelines Board of Directors has appointed Michael J. Parini as CEO and Executive Director of the Company, effective immediately, said Dr. Chris Hollowood, Chairman of Freeline. Michael has a long track record of building strong executive leadership teams and has had a significant positive impact on Freeline since he joined the Company from Vertex Pharmaceuticals. In a short period of time, he has mobilized program execution across our pipeline, increased focus on our core scientific and platform technology capabilities, and driven efficiencies across the organization that have extended our cash runway. In doing so, Michael has demonstrated his ability to unlock value at Freeline and deliver on the promise of its therapies and science to patients.

Dr. Hollowood continued, I wish to thank Theresa for leading the Company through the successful completion of its initial public offering last year and the extraordinary challenges of the COVID-19 pandemic. We wish her well in her future endeavors.

Freeline is at the forefront of gene therapy, leveraging a platform innovation engine that holds the potential to deliver functional cures to patients who suffer from debilitating diseases, said Mr. Parini, CEO of Freeline. I joined Freeline to deliver on the differentiated promise of our pipeline and technology, and am honored and excited to take the helm at this critical time for the Company.

Mr. Parini continued, I see three immediate strategic priorities as we turn the page on a new chapter for the Company. First and foremost is our commitment to advancing our pipeline with urgency for patients. Our recent execution in the clinic is a testament to this commitment, and we remain on track to deliver all expected data and developmental milestones across our four clinical and pre-clinical stage gene therapy programs before year end.

Our second strategic priority is to drive increased focus, financial discipline and operational efficiency in our business. Based on a thorough review of our operations, we have identified initial savings to extend our cash runway by nearly two quarters into the first quarter of 2023, funding operations through multiple value-generating milestones. We will continue this work to optimize our business, operational and platform strategy.

Finally, our third priority is to unlock the full value in our proprietary scientific and platform technology. We are refreshing our scientific strategy to leverage our unique combination of manufacturing quality, capsid potency and protein engineering capabilities, which we believe can drive lower and safer doses for high therapeutic effect, into the next wave of pipeline programs. We plan to focus our research and platform efforts where our differentiated capabilities allow us to address unmet needs and create transformative treatments for patients.

Mr. Parini added, We believe gene therapy holds tremendous promise. With our three strategic priorities in place, I look forward to building a world-class team and organization to realize the full potential of Freeline.

Key Pipeline and Operational Updates

Hemophilia B

Fabry Disease

Gaucher Disease (Type 1)

Hemophilia A

Platform Technology

Corporate

Q2 2021 Financial Highlights

Cash Position: Cash and cash equivalents were $164.7million as of June 30, 2021, as compared to $230.0million as of December 31, 2020. Based on the Companys revised operating plan, Freeline expects that its current level of cash and cash equivalents will enable the Company to fund its operating expenses into the first quarter of 2023.

R&D Expenses: Research and development (R&D) expenses for the six months ended June 30, 2021 were $48.1million, as compared to $29.4million for the same period in 2020. The increase of $18.7million was driven by increased investment in activities related to the current and proposed clinical trials for FLT201 and FLT210 and overall research and development, which includes earlier pipeline programs and further development of the Freeline platform.

G&A Expenses: General and administrative (G&A) expenses for the six months ended June 30, 2021 were $24.6million, as compared to $11.7million for the same period in 2020. The increase of $12.9million was driven primarily by an increase in personnel expenses, primarily due to an increase in headcount in corporate, legal, general and administrative functions to support the Companys growth initiatives and public company requirements, as well as an increase in non-cash share-based compensation expense, primarily due to equity grants to employees related to the completion of the Series C financing and the IPO.

As of June 30, 2021, the Company had 35,802,840 ordinary shares outstanding.

About Michael J. Parini

Michael Parini joined Freeline in March of 2021 to serve as the Companys President and Chief Operating Officer. Before coming to Freeline, Mr. Parini was the Executive Vice President and Chief Administrative, Legal and Business Development Officer at Vertex Pharmaceuticals, Inc. from March 2020 until February 2021, among other roles he held in his over five years with the company. Prior to Vertex, Michael Parini spent over a decade at Pfizer Inc. serving in multiple leadership roles within the company's global legal team, including Senior Vice President and Associate General Counsel.

About Freeline Therapeutics

Freeline is a clinical-stage biotechnology company developing transformative adeno-associated virus (AAV) vector-mediated systemic gene therapies. The Company is dedicated to improving patient lives through innovative, one-time treatments that provide functional cures for inherited systemic debilitating diseases. Freeline uses its proprietary, rationally-designed AAV vector, along with novel promoters and transgenes, to deliver a functional copy of a therapeutic gene into human liver cells, thereby expressing a persistent functional level of the missing protein into the patients bloodstream. The Companys integrated gene therapy platform includes in-house capabilities in research, clinical development, manufacturing and commercialization. The Company has clinical programs in HemophiliaB and Fabry disease, as well as preclinical programs in Gaucher disease Type 1 and HemophiliaA. Freeline is headquartered in the UK and has operations in Germany and the US.

Forward-Looking Statements

This press release contains statements that constitute forward looking statements as that term is defined in the United States Private Securities Litigation Reform Act of 1995, including statements that express the Companys opinions, expectations, beliefs, plans, objectives, assumptions or projections regarding future events or future results, in contrast with statements that reflect historical facts. Examples include, among other topics, discussion of the Companys strategies, anticipated operating and financial performance and financial condition, including its review of strategy and operations; the Companys expectations regarding its use of cash and cash runway; statements regarding the initiation, timing, progress and results of the Companys preclinical studies and clinical trials, including the initiation and full enrollment of the Phase1/2 dose confirmation trial for FLT180a and data readouts from that trial, progress with respect to the dose-escalation for the Phase1/2 dose-finding clinical trial of FLT190 and data readouts from that trial, commencement of the Phase 1/2 dose-finding clinical trial of FLT201, and completion of pre-clinical IND/CTA-enabling studies of FLT210; and manufacturing, research, pipeline, and clinical trial plans, including anticipated clinical development milestones for the Companys product candidates. In some cases, you can identify such forward-looking statements by terminology such as anticipate, intend, believe, estimate, plan, seek, project or expect, may, will, would, could or should, the negative of these terms or similar expressions. Forward looking statements are based on managements current beliefs and assumptions and on information currently available to the Company, and you should not place undue reliance on such statements. Forward-looking statements are subject to many risks and uncertainties, including the Companys recurring losses from operations; the uncertainties inherent in research and development of the Companys product candidates, including statements regarding the timing of initiation, completion and the outcome of clinical studies or trials and related preparatory work and regulatory review, regulatory submission dates, regulatory approval dates and/or launch dates, as well as risks associated with preclinical and clinical data, including the possibility of unfavorable new preclinical, clinical or safety data and further analyses of existing preclinical, clinical or safety data; the Companys ability to design and implement successful clinical trials for its product candidates; the recent departures of a number of executive officers of the Company, and the Companys ability to fill open positions, implement an orderly transition process and retain key talent; whether the Companys cash resources will be sufficient to fund the Companys foreseeable and unforeseeable operating expenses and capital expenditure requirements for the Companys expected timeline; the potential for a pandemic, epidemic or outbreak of infectious diseases in the US, UK or EU, including the COVID-19 pandemic, to disrupt and delay the Companys clinical trial pipeline; the Companys failure to demonstrate the safety and efficacy of its product candidates; the fact that results obtained in earlier stage clinical testing may not be indicative of results in future clinical trials; the Companys ability to enroll patients in clinical trials for its product candidates; the possibility that one or more of the Companys product candidates may cause serious adverse, undesirable or unacceptable side effects or have other properties that could delay or prevent their regulatory approval or limit their commercial potential; the Companys ability to obtain and maintain regulatory approval of its product candidates; the Companys limited manufacturing experience which could result in delays in the development, regulatory approval or commercialization of its product candidates; and the Companys ability to identify or discover additional product candidates, or failure to capitalize on programs or product candidates. Such risks and uncertainties may cause the statements to be inaccurate and readers are cautioned not to place undue reliance on such statements. We cannot guarantee that any forward-looking statement will be realized. Should known or unknown risks or uncertainties materialize or should underlying assumptions prove inaccurate, actual results could vary materially from past results and those anticipated, estimated or projected. Investors are cautioned not to put undue reliance on forward-looking statements. A further list and description of risks, uncertainties and other matters can be found in the Companys Annual Report on Form 20-F for the fiscal year ended December 31, 2020 and in subsequent reports on Form 6-K, in each case including in the sections thereof captioned Cautionary Statement Regarding Forward-Looking Statements and Item 3.D. Risk factors. Many of these risks are outside of the Companys control and could cause its actual results to differ materially from those it thought would occur. The forward-looking statements included in this press release are made only as of the date hereof. The Company does not undertake, and specifically declines, any obligation to update any such statements or to publicly announce the results of any revisions to any such statements to reflect future events or developments, except as required by law. For further information, please reference the Companys reports and documents filed with theU.S. Securities and Exchange Commission (the SEC). You may review these documents by visiting EDGAR on theSECwebsite atwww.sec.gov.

Contact

David S. ArringtonVice President Investor Relations & Corporate CommunicationsFreeline Therapeuticsdavid.arrington@freeline.life+1 (646) 668 6947

Freeline Therapeutics Holdings plcUnaudited Condensed Consolidated Statements of Operations Data(in thousands of U.S. dollars, except per share data)

Freeline Therapeutics Holdings plcUnaudited Condensed Consolidated Balance Sheet Data(in thousands of U.S. dollars, except per share data)

See more here:
Freeline Appoints Michael J. Parini as Chief Executive Officer and Reports Second Quarter 2021 Financial Results - GlobeNewswire

Posted in Gene therapy | Comments Off on Freeline Appoints Michael J. Parini as Chief Executive Officer and Reports Second Quarter 2021 Financial Results – GlobeNewswire

Scientists Grew Stem Cell ‘Mini Brains’. Then, The Brains Sort-of Developed Eyes – ScienceAlert

Posted: August 18, 2021 at 2:00 am

Mini brains grown in a lab from stem cells have spontaneously developed rudimentary eye structures, scientists report in a fascinating new paper.

On tiny, human-derived brain organoids grown in dishes, two bilaterally symmetrical optic cups were seen to grow, mirroring the development of eye structures in human embryos. This incredible result will help us to better understand the process of eye differentiation and development, as well as eye diseases.

"Our work highlights the remarkable ability of brain organoids to generate primitive sensory structures that are light sensitive and harbor cell types similar to those found in the body," said neuroscientist Jay Gopalakrishnan of University Hospital Dusseldorf in Germany.

"These organoids can help to study brain-eye interactions during embryo development, model congenital retinal disorders, and generate patient-specific retinal cell types for personalized drug testing and transplantation therapies."

(Elke Gabriel)

Brain organoids are not true brains, as you might be thinking of them. They are small, three-dimensional structures grown from induced pluripotent stem cells - cells harvested from adult humans and reverse engineered into stem cells, that have the potential to grow into many different types of tissue.

In this case, these stem cells are coaxed to grow into blobs of brain tissue, without anything resembling thoughts, emotions, or consciousness. Such 'mini brains' are used for research purposes where using actual living brains would be impossible, or at the very least, ethically tricky - testing drug responses, for example, or observing cell development under certain adverse conditions.

This time, Gopalakrishnan and his colleagues were seeking to observe eye development.

In previous research, other scientists had used embryonic stem cells to grow optic cups, the structures that develop into almost the entire globe of the eye during embryonic development. And other research had developed optic cup-like structures from induced pluripotent stem cells.

Rather than grow these structures directly, Gopalakrishnan's team wanted to see if they could be grown as an integrated part of brain organoids. This would add the benefit of seeing how the two types of tissue can grow together, rather than just growing optic structures in isolation.

"Eye development is a complex process, and understanding it could allow underpinning the molecular basis of early retinal diseases," the researchers wrote in their paper.

"Thus, it is crucial to study optic vesicles that are the primordium of the eye whose proximal end is attached to the forebrain, essential for proper eye formation."

Previous work in the development of organoids showed evidence of retinal cells, but these did not develop optic structures, so the team changed their protocols. They didn't attempt to force the development of purely neural cells at the early stages of neural differentiation, and added retinol acetate to the culture medium as an aid to eye development.

(Gabriel et al., Cell Stem Cell, 2021)

Their carefully tended baby brains formed optic cups as early as 30 days into development, with the structures clearly visible at 50 days. This is consistent with the timing of eye development in the human embryo, which means these organoids could be useful for studying the intricacies of this process.

There are other implications, too. The optic cups contained different retinal cell types, which organized into neural networks that responded to light, and even contained lens and corneal tissue. Finally, the structures displayed retinal connectivity to regions of the brain tissue.

"In the mammalian brain, nerve fibers of retinal ganglion cells reach out to connect with their brain targets, an aspect that has never before been shown in an in vitro system," Gopalakrishnan said.

And it's reproducible. Of the 314 brain organoids the team grew, 73 percent developed optic cups. The team hopes to develop strategies for keeping these structures viable on longer time-scales for performing more in-depth research with huge potential, the researchers said.

"Optic vesicle-containing brain organoids displaying highly specialized neuronal cell types can be developed, paving the way to generate personalized organoids and retinal pigment epithelial sheets for transplantation," they wrote in their paper.

"We believe that [these] are next-generation organoids helping to model retinopathies that emerge from early neurodevelopmental disorders."

The research has been published in Cell Stem Cell.

Continue reading here:
Scientists Grew Stem Cell 'Mini Brains'. Then, The Brains Sort-of Developed Eyes - ScienceAlert

Posted in Stem Cells | Comments Off on Scientists Grew Stem Cell ‘Mini Brains’. Then, The Brains Sort-of Developed Eyes – ScienceAlert

Cell therapy strategies for COVID-19: Current approaches and potential applications – Science Advances

Posted: August 18, 2021 at 2:00 am

Abstract

Coronavirus disease 2019 (COVID-19) continues to burden society worldwide. Despite most patients having a mild course, severe presentations have limited treatment options. COVID-19 manifestations extend beyond the lungs and may affect the cardiovascular, nervous, and other organ systems. Current treatments are nonspecific and do not address potential long-term consequences such as pulmonary fibrosis, demyelination, and ischemic organ damage. Cell therapies offer great potential in treating severe COVID-19 presentations due to their customizability and regenerative function. This review summarizes COVID-19 pathogenesis, respective areas where cell therapies have potential, and the ongoing 89 cell therapy trials in COVID-19 as of 1 January 2021.

Coronavirus disease 2019 (COVID-19) continues to strain patients, providers, and health care systems worldwide. Since its discovery, the disease has contributed to approximately 200 million infections and 4 million deaths worldwide. The scientific community has focused vast resources on understanding the virus causing COVID-19, named severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and the pathologies associated with the infection. Enormous effort has been placed to shed light on the mechanisms of viral entry and infection, the interaction between the virus and the hosts immune system, and the mechanisms of injury underlying the common manifestations of the disease.

SARS-CoV-2 initially emerged as a pathogen causing mainly viral pneumonias; however, experience in the proceeding months showed that the disease manifests throughout the body, leading to pathologies of the immune, renal, cardiac, and nervous systems, among others. While most patients have a mild course, over 15% develop severe and critical disease (1), leading to a substantial number of patients requiring prolonged hospitalization with intensive care services and potentially facing subsequent chronic manifestations related to pathological injuries from the disease process. In addition, mortality can be as high as 61.5% in critically ill patients with the disease (2).

As we begin to appreciate the subacute and chronic sequela of COVID-19, it is crucial to focus research efforts on finding therapies that not only dampen the acute damage but also can do so in a targeted manner while restoring physiological function and addressing the long-term sequela of the disease. Cell therapies have the potential to regenerate damaged tissue and tackle the immune system and, hence, are a treatment option with great promise. Here, we provide an overview of the COVID-19 pathogenesis in various organ systems, the overall advantages of cell therapies, potential cell targets and strategies within each organ system, and a summary of current cell therapy studies and trials for COVID-19 as of 1 January 2021.

SARS-CoV-2 first interacts with cells via binding of the viral spike protein to angiotensin-converting enzyme 2 (ACE2) on the cell surface (3, 4). After binding to ACE2, the spike protein is processed by the host transmembrane protease serine 2 (TMPRSS2), priming it for membrane fusion. This is considered to be the primary route of infection in vivo. Alternatively, the virus can be taken up into the cell via endocytosis and the spike protein processed by the endosomal proteases cathepsins B and L (3). After fusion with the cell membrane and release into the cytoplasm, the RNA replication machinery encoded in the first open reading frame of the viral genome is translated, followed by RNA replication and viral protein translation. SARS-CoV-2 co-opts and alters numerous cellular proteins and pathways, many of which are yet to be elucidated (5). It has been indicated that neuropilin 1 (NRP1) has a role in potentiating SARS-CoV-2 entry through the ACE2 pathway (6, 7). Studies from other coronaviruses provided evidence for CD147 and the 78-kDa glucose-regulated protein (GRP78) as putative alternative receptors, but more investigations on how the collective tissue distribution of these factors correlate with viral tropism and disease symptoms are under active investigation (8, 9).

Cellular tropism of SARS-CoV-2 is considered to be largely dictated by the distribution of ACE2. Bulk transcriptomic studies found ACE2 primarily expressed in the lungs, intestinal tract, kidneys, gallbladder, and heart; lower levels of expression were observed in the brain, thyroid, adipose tissue, epididymis, ductus deferens, breast, pancreas, rectum, ovary, esophagus, liver, seminal vesicle, salivary gland, placenta, vagina, lung, appendix, and skeletal muscle (1012). In the respiratory tract, ACE2 is most highly expressed in nasal epithelial cells, where SARS-CoV-2 is thought to initially infect followed by propagation into the distal alveoli (13). Many organs that express higher levels of ACE2 are not major sites of viral replication, indicating that expression of other host factors, including TMPRSS2, NRP1, and host restriction factors likely contributes to viral tropism (12).

Although most patients infected with SARS-CoV-2 present with mild symptoms (14), a considerable part of the population, including elderly patients and those with underlying comorbidities, have an increased risk of more severe outcomes, including death (15). Current treatment options for severely ill patients, aimed at reducing inflammation during the acute phase of the infection, have their limitations. Medications may be nonspecific for SARS-CoV-2 targets or are repurposed without a clear mechanism of benefit, while others such as remdesivir and tocilizumab may not be readily accessible because of federal allocations or cost barriers (16). In addition, these treatments have not focused on long-term sequela of the disease such as regeneration of damaged tissue structure and function. Cell therapies may thus be a promising class of therapies that could overcome these challenges through their customizability, targetability, scalable manufacturing, and restoration of function.

Cellular therapies have shown success in treating conditions that have otherwise been challenging to manage with mainstream treatment modalities, including, but not limited to, oncologic, neurodegenerative, and immunologic disorders. Cell therapy approaches including, but not limited to, mesenchymal stromal cells (MSCs), induced pluripotent stem cells (iPSCs), and T cells have been widely studied, and their efficacy has led to several U.S. Food and Drug Administration (FDA) approvals of cell therapies including, most famously, axicabtagene ciloleucel (Yescarta) and tisagenlecleucel (Kymriah) (1720). Extensive safety and efficacy data from cell therapies trials in various indications suggest that cell therapies could play a role in treating patients with COVID-19 as well.

Two potential concerns with cell therapies are immune rejection and tumorigenicity. Immune rejection concerns for allogeneic cell therapy have been discussed in the literature, especially as new cell therapies emerge. MSCs, for example, are considered to be immune suppressive and immune evasive, yet, the standard of treatment using allogeneic MSCs is the addition of immunosuppressive regimens alongside the cell therapy (21, 22). While immunosuppressive therapy may be used to protect the graft, it may not always prevent graft rejection and can come with its own adverse effects. Genome engineering can help address the immune system by tackling both the innate and adaptive immune systems. Potential strategies include knocking out genes responsible for immune system activation, such as major histocompatibility complex I and II (23, 24). These modifications could address both the acute and chronic rejection phases, making the cell grafts more resistant to the host immune system.

Tumorigenicity is an important consideration with cell therapies. The risk of tumorigenicity seems to be greater with MSCs, iPSCs, and human embryonic stem cells (hESCs), and it can present in the form of teratoma or as a true tumor (2527). This risk can be reduced by increasing the efficiency of differentiation to the target cell type thereby reducing residual pluripotent cells, such as by transcription factormediated cell programming or by incorporating suicide genes into cell grafts that can be activated in the rare chance a graft becomes malignant (2830). Several suicide mechanisms have been described in the literature, including a recent study by Itakura et al. (31) in which iCaspase9 was inserted as a fail-safe system in iPSC cell lines. If these cell lines become cancerous once transplanted in mice, induction of the iCaspase9 with a small molecule showed the formed tumors to rapidly reduce in size (31). These approaches increase the safety profile of cell therapies for clinical applications in patients with COVID-19 and beyond.

A clear understanding of COVID-19 pathogenesis is necessary to appreciate the potential benefit of cell therapies. Cell therapies provide paramount benefit as potential targeted treatment strategies to address localized damage inflicted by the disease and restore physiological functions (Fig. 1). In 2020, March and April recorded a large initial surge in global COVID-19 cases and deaths, as presented by the World Health Organization. There was a concurrent increase in the numbers of cell therapybased clinical trials initiated during those 2 months (Fig. 2A). As of 1 January 2021, there are 89 cell therapybased clinical trials registered on clinicaltrials.gov (Table 1) targeting COVID-19 pathology. Most of the clinical trials are held in the United States and China, 36% and 16%, respectively, with the rest of the clinical trials spread across the globe (Fig. 1B). MSCs constitute the majority cell type used in the cell therapy clinical trials, around 71%, with the rest using cell types such as natural killer (NK) cells, T cells, early apoptotic cells, and others (Fig. 1C). About 88% of the clinical trials are in phases 1 and 2, with one trial in phase 2/3 and one in phase 3 (Fig. 2D). The enrollment in each clinical trial was most frequently 21 to 30 patients but ranged up to 400 depending on the phase of the trial (Fig. 2E). In addition, the variability of patient enrollment numbers could be due to the varying statuses of each clinical trial (Fig. 2F). It is also worth noting that over half of the cell therapybased clinical trials are sponsored and supported by the industry sector (Fig. 2G), which indicates the pivotal role for industry in accelerating the necessary research to combat COVID-19.

Blue text boxes describe specific pathogenesis for each organ system. Green text boxes describe potential and ongoing cell therapy applications for each organ system. ALT, alanine aminotransferase; AST, aspartate aminotransferase.

(A) Number of COVID-19 targeting cell therapy clinical trials started in each month of the year 2020. (B) World map showing global distribution of the registered cell therapy clinical trials and their numbers per country. (C) Different cell types used in the cell therapybased clinical trials and their respective count. (D) Stages of the 89 cell therapy clinical trials registered as of 1 January 2021. (E) Distribution of patient enrollment numbers across the 89 clinical trials. (F) Breakdown of the 89 cell therapy clinical trial statuses. (G) The percentages of cell therapies sponsored and supported by the industry sector.

Search approach: performed 1 January 2021; Clinicaltrials.gov: advanced search; Condition - OVID; Study Type -Interventinal; Intervention/treatment - Cell; of 157 studies, exclude nonCOVID-19 patients (n = 12) and noncell therapy trials (n = 56); leaving 89 available studies. NCT, national clinical trial.

Pulmonary symptoms are the mainstay of COVID-19 and may include dry cough, dyspnea, pneumonia, and acute respiratory distress syndrome (ARDS) (32). Bilateral pulmonary infiltrates and ground-glass opacities are seen radiographically in over 70% of hospitalized patients (14). Furthermore, ARDS has shown to be present in over 90% of deceased patients (33). ARDS and the associated alveolar damage are thought to be primarily due to immune-related response (3, 34). Other pulmonary complications may include secondary pulmonary hypertension, hypercoagulability-related pulmonary emboli, and long-lasting fibrosis in patients who do recover from the acute infection (35, 36).

Some preclinical data suggest that patients with COVID-19 may benefit from cell therapies, particularly using MSCs in models of viral and inflammatory lung damage (37). For instance, MSCs were found to reduce the impairment of alveolar fluid clearance caused by influenza A H5N1 infection in vitro and mitigate lung injury in vivo (38). Another study showed that MSC treatment reduces influenza H9N2induced acute lung injury in mice and reduces pulmonary inflammation (39). In another study, MSCs were shown to promote macrophages to become anti-inflammatory and take on a phagocytic phenotype through extracellular vesicles, thereby ameliorating lung injury in mice (40).

Several studies have described promising treatment of pneumonia and ARDS in critically ill patients with COVID-19 using cell therapies. In China, Liang et al. (41) reported treatment of one patient with severe COVID-19 unresponsive to steroid medications, after three successive injections of 5 107 human umbilical cord MSCs at days 1, 4, and 7 of treatment initiation. The patients pulmonary lesions had begun to resolve by day 7 after the first MSC injection. Tang et al. (42) reported treatment with allogeneic menstrual bloodderived MSCs of two patients with COVID-19 with ARDS. Treatment involved three successive injections of 1 106 MSCs/kg of body weight at days 1, 2, and 4 of treatment initiation. Both patients were discharged from the hospital. Leng et al. (43) reported a pilot study where they transplanted a single dose of 1 106 MSCs/kg of body weight in seven patients with mild, severe, and critical COVID-19, with three patients on the placebo arm. Results from the study showed overall safety of the treatment, with two severe patients recovering and being discharged within 10 days of treatment. In Spain, Sanchez-Guijo et al. (44) treated 10 patients under mechanical invasive intubation with either one, two, or three doses of 1 106 adipose-derived MSCs/kg of body weight. Seven of the 13 patients were extubated approximately 7 days after initiation of treatment. Furthermore, the authors observed that patients who received cell therapy earlier in their disease course had better outcomes. These open labeluncontrolled administrations are important as they demonstrate apparent safety with no obvious adverse events.

Various MSC-based strategies are assessing treatment of patients with COVID-19 with pulmonary symptoms, especially pneumonia and ARDS. One phase 1/2a randomized double-blind trial (NCT04355728) assessed administration of two infusions of 1 107 umbilical cordderived MSCs for COVID-19 ARDS, showing improved 28-day survival following therapy (91% in treatment group, n = 12 versus 42% in control, n = 12) (45). Another phase 3 study comparing administration of two injections of 2 106 MSCs/kg of body weight and standard of care compared to placebo injection and standard of care in patients with COVID-19 with moderate to severe ARDS failed to meet the primary end point of 43% reduction in mortality in an interim analysis (NCT04371393). Thus, further investigation is necessary to determine whether MSC-based therapy could improve COVID-related lung injury.

COVID-19related lung fibrosis has been characterized by fibroblast proliferation, airspace obliteration, and microhoneycombing, which is thought to persist in patients who survive the acute infection (46). This pattern of fibrotic change may be similar to that of idiopathic pulmonary fibrosis (IPF) (36), and prior cell therapy studies in IPF may shed light on potential avenues for cell therapy applications in patients with COVID-19. IPF is a progressive disease of unknown etiology that leads to fibrosis of the lungs and is the primary cause of more than half of all lung transplants worldwide (47). Cell therapies using type II pneumocytes (PTIIs), which are progenitors of the lung alveolar epithelium, have shown efficacy in preclinical animal models of IPF by regenerating lung epithelium, releasing surfactant, and reversing pulmonary fibrosis (48, 49). A phase 1 clinical study also showed that targeted intratracheal delivery of PTIIs showed safety and clinical stability at 12-month follow-up of 16 patients with moderate to severe IPF (50). In addition to PTIIs, MSCs have also been used in IPF. A recent randomized trial of patients with IPF treated with two doses of 2 108 allogenic bone marrow MSCs every 3 months for 1 year showed safety and improved respiratory function when compared to control participants (51). This suggests that even patients with COVID-19 with residual chronic fibrosis may benefit from cell-based therapies in the future, although further data are necessary to support this conclusion. Ultimately, cell therapies that can reverse fibrotic changes or supplement normal pneumocyte function could address potential chronic pulmonary effects from COVID-19.

The hosts immune response toward SARS-CoV-2 has been studied carefully since the outbreak, with many potential mechanisms of interaction being elucidated on the basis of similarities of the virus to SARS-CoV. Most patients with COVID-19 mount antibody responses to SARS-CoV-2, which vary in magnitude and potency (52). Neutralizing antibodies appear to target the receptor binding domain of the spike proteins (52, 53). Patients with high immunoglobulin M (IgM) and immunoglobulin G (IgG) titers have a worse prognosis (54), which could be correlated with high viral load but could also indicate a harmful robust immune response through antibody-dependent enhancement (ADE). ADE is a phenomenon that has been observed in several viruses, including SARS-CoV, where viral-specific antibodies promote viral entry into immune cells expressing Fc receptors (55), such as monocytes, macrophages, and B cells, leading to enhanced amplification of the virus. Implications of ADE in COVID-19 have been discussed in greater detail by Eroshenko et al. (56). With regard to T cells, several studies have compared leukocyte profiles between patients with mild and severe manifestations of the disease and showed decreased T cell count in both CD4+ and CD8+ populations, more commonly in intensive care unit (ICU) patients but highly prevalent in non-ICU patients as well (57). Lower levels of CD4+ T helper cells and CD8+ cytotoxic T cells likely hinder the ability of the immune system to neutralize and kill viral-infected cells.

In addition, a marked increase of proinflammatory cytokines such as interleukin-1 (IL-1), IL-6, tumor necrosis factor (TNF-), and interferon- (IFN-) has been observed in patients with severe COVID-19 (57, 58). In these cases, SARS-CoV-2 immune evasion leads to a robust viral replication and a delayed and dysregulated IFN- response, resulting in recruitment and accumulation of inflammatory macrophages and neutrophils (58, 59). Further IFN- activation by these cells leads to additional cytokine and chemokine signals [IFN-, TNF-, C-C motif chemokine ligand (CCL)2, CCL7, and CCL12] that enhance infiltration and activation of monocytes and neutrophils, further exacerbating the inflammatory response and inducing high cytokine levels, a phenomenon referred to as cytokine storm, which has been linked to more severe manifestations of COVID-19 (60).

Several immune-based cell strategies can be proposed for targeting different pathologies of COVID-19. Several NK cell therapies for COVID-19 are under investigation (Table 1). NK cells are activated and recruited at the site of infection in response to IL-12 and IL-18 signals. They control viral replication using perforin and granzyme granules and induce Fas ligand or TNF-arelated apoptosis-inducing ligandmediated apoptosis in infected cells (61). Cell therapies involving NK cells and chimeric antigen receptor (CAR) NK cells have shown clinical safety and efficacy in numerous oncological indications (62), and they may have a role in treating various infectious pathologies as well (63). As NK cells recognize viral infected cells by identifying up-regulated stress markers and down-regulated inhibitory ligands, exogenous administration of NK cellbased therapies could thus assist in identifying SARS-CoV-2infected cells and promote viral clearance (64). A phase 1 study is assessing the efficacy and safety of CYNK-001 cells, which are allogeneic, off-the-shelf, and cryopreserved NK cells derived from CD34+ human placental stem cells, in 14 adult patients with mild to moderate COVID-19 (NCT04365101). In another phase 1 study, FT516 cells, which are allogeneic, off-the-shelf, and cryopreserved NK cells derived from iPSCs, are being tested for efficacy and safety in 12 adult patients with COVID-19 who are hospitalized and fulfill requirements for hypoxia (NCT04363346). With regard to CAR NK cells, a phase 1/2 study in China is using off-the-shelf NKG2D-ACE2 CAR NK cells to target viral infected cells while also secreting IL-15 as a superagonist and granulocyte-macrophage colony-stimulating factor neutralizing single-chain variable fragment to reduce the likelihood of cytokine release syndrome (NCT04324996). Intravenous infusion of 1 108 cells/kg of body weight will be administered weekly in patients with COVID-19, and the study is currently recruiting patients.

Given that immune system overactivation is a significant factor in pathologies of COVID-19, another potential strategy could involve CD4+CD25+Foxp3+ regulatory T cells (Tregs). Tregs function by secreting anti-inflammatory cytokines IL-10 and transforming growth factor (TGF-) as well as by contact-dependent signaling, and have been shown to inhibit the influx of neutrophils to the lung, induce apoptotic cell clearance of activated neutrophils and macrophages, and decrease proinflammatory cytokine levels (65, 66). Moreover, they can inhibit excessive innate immune responses via induction of secondary immunosuppressive neutrophils that generate anti-inflammatory cytokines and via enzymes indoleamine 2,3-dioxygenase and heme oxygenase-1, which further inhibit cellular proliferation (66). The safety and feasibility of Tregs has been clinically evaluated over the past decade, showing tolerability and clinical improvement especially in the setting of solid-organ transplantation and autoimmune disease (67). Hence, the immunosuppressive role of Tregs may be beneficial in quelling the cytokine storm in patients with COVID-19. Potential strategies may include using polyclonal expanded Tregs versus engineered antigen-specific Treg approaches. Polyclonal Tregs offer a more generalized immunosuppressive strategy, which may be similar to current immunosuppressive medications. Polyclonal Tregs have been clinically evaluated with promising results in type 1 diabetes and other autoimmune diseases (68), but they have not been clinically tested in immune overactivation in viral infections. A concern with this therapy would be the exacerbation of acute infection by excessive quelling of the host immune response to SARS-CoV-2. Engineered antigen-specific Tregs could help localize immunosuppressive effects (65), but this could also facilitate enhanced viral replication. Overall, Treg therapies could aid in suppressing the overactive immune system in patients with COVID-19 (69), but generalizing early safety data from clinical trials of autoimmune and transplant patients toward patients with COVID-19 would need careful evaluation. Two phase 1 clinical trials, which are not yet recruiting, are aiming to test the efficacy and safety of allogeneic, off-the-shelf, and cryopreserved Treg cell infusions in patients with COVID-19 with moderate to severe ARDS (NCT04468971) or intubated and mechanically ventilated (NCT04482699).

Besides Tregs, other T cell therapies are being evaluated for COVID-19 (Table 1). Viral-specific T cells are currently under investigation in three trials, and they are using viral-specific T cells from healthy donors who have mounted an appropriate response to the SARS-CoV-2 (NCT04457726, NCT04406064, and NCT04401410). A better understanding of effective targets could aid in the development of engineered T cells from more accessible and scalable sources than previously infected healthy donors. In addition, a phase 1/2 trial evaluating the use of RAPA-501, a hybrid T helper 2/Treg phenotype, aims to suppress immune overactivation in a T cell receptorindependent manner (NCT04482699). Engineered T cells, particularly CAR T therapies, have shown promise in the treatment of immune system overactivation in diseases such as pemphigus vulgaris, type 1 diabetes, and lupus (70), and targeted T cell therapies could play a role in treating COVID-19 immune overactivation and facilitating viral clearance. Recent single-cell sequencing studies of patients with COVID-19 have shown an increase in monocytes, macrophages, and clonally expanded CD8+ T cells, which may contribute to the cytokine storm seen in severe cases (71, 72). This provides a rationale to direct cell therapies such as CAR T/NK cells to target these enriched populations with the goal of reducing the excess cell population, and potentially decreasing the severity of the cytokine storm. In addition, B lymphocytes could theoretically be engineered to recombinantly express humanized monoclonal antibodies with neutralizing antiSARS-CoV-2 activity. However, convalescent plasma or monoclonal antibodies likely have similar benefits without the increased complexity of a cell therapybased modality (73).

In addition to their role in targeting COVID-19related lung damage, MSCs are also an intriguing target for immune-based cell therapy because of their immunomodulatory capacities. In the lung, MSCs mediate immune homeostasis by TNF- and IL-1induced up-regulation of anti-inflammatory cytokines such as protein TNF-stimulated gene 6, IL-10, TGF-, prostaglandin E2, and nitric oxide (74, 75). Moreover, by modulating overactivation of the immune system, MSCs have shown efficacy for the treatment of immune-related conditions such as steroid-refractory graft-versus-host disease and systemic lupus erythematosus (76, 77). Hence, MSC therapy may play a role in suppressing COVID-19associated immune activation and cytokine storm. Several recent studies have reported decreases in inflammatory marker levels after treatment with MSCs that correlated with clinical improvement (4144). Moreover, ongoing clinical trials are assessing the immunomodulatory capabilities of MSCs in patients with COVID-19 (NCT04348435, NCT04377334, and NCT04397796). Another phase 1 clinical trial is assessing the efficacy and safety of allogeneic umbilical cord bloodderived MSCs as adjuvant therapy in patients receiving oseltamivir and azithromycin (NCT04457609). Dosing for MSC trials varies widely between 5 105 and 1 107 cells/kg or 2 107 and 2 108 cells per dose with the number of doses ranging from one to four. Cell sourcing for MSC trials includes the umbilical cord, placenta, adipose tissue, intra-aortic tissue, olfactory mucosa, and the dental pulp (78). More detailed reviews on mechanisms of MSC immunomodulation and potential benefits in COVID-19 have been previously explored (75, 7889).

Neurological manifestations are a significant consideration in patients with COVID-19 and are reported in 57.4% of confirmed cases (90). Presenting symptoms range from headache, anosmia, and ageusia to more serious manifestations such as ischemic stroke, encephalitis, and encephalomyelitis (91). The innate immune response is likely responsible for symptoms such as headache and encephalitis through uncontrolled cytokine release. However, symptoms such as anosmia, encephalomyelitis, and stroke suggest potential viral invasion of the central nervous system (CNS). Proposed mechanisms of CNS viral access include retrograde axonal transport through vagal afferents peripherally (92) or via direct CNS invasion, as studies have shown ACE2 receptors to be expressed in several regions of the brain, especially in oligodendrocytes and astrocytes (93). The symptoms of anosmia and ageusia were initially suggestive of CNS invasion, especially as SARS-COV studies had shown that the virus could enter the brain through the olfactory nerve within days of infection, causing inflammation and demyelination (94). However, analysis of human RNA sequencing and single-cell sequencing data showed that ACE2 and TMPRSS2 are not expressed in olfactory sensory nerves but instead in olfactory epithelium (95). Acute cerebral ischemic events have been reported in patients with COVID-19, especially in younger patients without typical risks of cerebrovascular disease (96, 97). These manifestations are likely due to an overall prothrombotic state, potential down-regulation of ACE2, which causes an overall loss of neuroprotection, and hyperinflammatory cytokine release. In addition, there has been an increasing number of reports of Guillain-Barre syndrome and its variants, transverse myelitis, and other demyelinating conditions in affected patients, some with multifocal lesions in the brain and spine (98). The presence of demyelination has also been present in autopsy studies (98). The etiology of these lesions is likely immune-related, potentially because of a delayed immune reaction.

To date, there have been no reports of cell-based clinical trials addressing neurologic manifestations in patients with COVID-19. However, the high incidence of neurologic manifestations coupled with increasing reports of demyelinating disease and ischemic stroke in affected patients may require treatment options that focus on long-term deficits, which can potentially be addressed via cell therapy. Regarding demyelination, oligodendrocyte precursor cells (OPCs) have been explored in the setting of spinal cord injury and have showed safety, tolerability, cell engraftment, and improved motor function at 12-month follow-up in patients (NCT02302157). In addition, human iPSC (hiPSC)derived OPCs were shown to remyelinate denuded axons in nonhuman primates with experimental autoimmune encephalomyelitis (EAE), a common animal model for multiple sclerosis (99). As COVID-19related demyelination is likely due to immune-mediated myelin damage, successful applications of OPCs in other demyelinating animal models such as EAE suggest a potential benefit of OPCs in COVID-19related refractory demyelination.

Patients with COVID-19 who suffered acute ischemic strokes, especially those with persistent deficits after mechanical thrombectomy or thrombolytic therapy, could also be a target of cell therapy. The long-term outcomes of patients suffering strokes, most of whom are younger and suffer large vessel occlusions, could be devastating. The prospect of stem cell therapies in stroke has expanded, with several concluded and ongoing clinical trials using bone marrowderived stem cells and neural stem cells (100). MASTERS-2 (NCT03545607) and TREASURE (NCT02961504) are ongoing phase 3 clinical trials assessing treatment outcomes after intravenous administration of bone marrowderived adult progenitor stem cells in patients suffering from ischemic stroke in the acute setting. Hence, this subpopulation of patients with COVID-19 may benefit from neuroregenerative cell therapies in the future.

Cardiac manifestations, such as elevated troponin levels and myocardial ischemic infarctions, are commonly seen in patients with COVID-19, particularly in severe presentations (101). Myocardial injury was found in 22% of hospitalized patients and nearly 60% of deceased patients (4, 14). Moreover, cardiac arrhythmias were shown to be present in 44% of patients with COVID-19 in the ICU (102). Although cardiac cells express high levels of ACE2 (11), it remains unclear whether these cases constitute direct or indirect injury. One study on hiPSC-derived cardiomyocytes from patients with COVID-19 suggests viral invasion and cytopathic features in cardiac tissue (103). As cell therapies are designed, one potential way to mitigate the risk of SARS-CoV-2 viral entry of the treatment may be to genetically modulate viral entry proteins within the cell therapy itself. Indirect injury could be due to systemic hypoxia, secondary pulmonary hypertension, arrhythmia due to metabolic derangements, and cytokine storm damage (104).

Early cell therapy trials in acute myocardial infarct have largely focused on bone marrow mononuclear cells (BMMNCs), and early studies such as BOOST (105) and TOPCARE-MI (106) showed improvements in infarct size and left ventricular ejection fraction. Subsequent trials such as BOOST-2 (107) and TIME (108) showed no clinical benefit, however, questioning the role of BMMNCs in acute myocardial infarction. Preclinical data using a combination of cardiopoietic stem cells and MSCs have been promising and are under investigation in human trials (NCT02501811) (109). Further, Menasch et al. (110) showed that hESC-derived cardiac progenitors given to six patients with ischemic left ventricular dysfunction showed clinical improvement in systolic function without new tumors or arrhythmias. Clinical applications of iPSC-derived cardiomyocytes are also being explored (111). These advances in cell-based cardiac therapy can potentially be exploited for patients suffering from COVID-19related cardiac ischemia. In addition, a recent clinical study used cardiosphere-derived cells, which are cardiac progenitor cells, to assess treatment of severe pulmonary manifestations in six patients with COVID-19. Four of the six patients were discharged from the hospital, while the remaining two were in stable conditions at the time the study was published (112). A phase 2 trial further assessing the efficacy of these cardiosphere-derived cells is currently under investigation (NCT04623671).

Gastrointestinal manifestations occur in 5 to 10% of COVID-19 cases; however, symptoms have been mild and self-limited to nausea, diarrhea, and vomiting, despite ACE2 and TMPRSS2 being coexpressed in the small and large intestines and SARS-CoV-2 being detected in fecal samples of infected patients, suggesting direct viral invasion of enterocytes (113). This suggests that chronic intestinal sequela is unlikely to occur, negating the need for advanced treatments such as cell therapy. Hepatic involvement also appears to be frequent. Elevations in alanine aminotransferase and aspartate aminotransferase have been reported in up to a third of patients (114). ACE2 expression has been identified in cholangiocytes (115, 116); however, histopathological examinations have yet to show direct viral inclusions in the liver (117). Other possibilities for hepatic injury may include immune-mediated damage, systemic hypoxia secondary to lung damage, and drug-induced liver injury (118). Stem cellderived hepatic cells have been studied in the setting of acute and chronic liver failure. Patients have received cell therapies through the portal vein or splenic artery with improvement in serological markers such as prothrombin time or severity of hepatic encephalopathy (119). Although hepatocyte-based therapies have largely been considered a bridge to transplantation rather than a curative therapy itself, rare cases of patients with COVID-19 with acute liver failure may benefit from hepatocyte-based therapies (120).

Renal manifestations are frequent and range from mild proteinuria to severe injury requiring renal replacement therapy (121). Pei et al. (122) showed that 75% of patients with COVID-19 presenting with pneumonia were found to have an abnormal urine dipstick. Moreover, the presence of acute kidney injury (AKI) was associated with increased mortality, and only 46% of those patients who developed an AKI showed complete resolution after 12 days of follow-up. Over 80% of AKIs were intrinsic, with the remainder being secondary to rhabdomyolysis; there were no cases of prerenal AKI (122). Pathological studies have demonstrated acute tubular necrosis, presence of microthrombi, and mild interstitial fibrosis; however, no evidence of lymphocytic infiltrate in affected patients was found (123). While direct viral invasion is possible as ACE2 expression is present in tubular epithelium and podocytes, secondary mechanisms appear more relevant in inducing renal damage, which may include systemic hypoxia, rhabdomyolysis, cytokine-mediated damage, microemboli due to hypercoagulability, and cardiorenal congestion from right heart strain (121).

Cellular therapies for kidney disease are currently being explored and may benefit patients with COVID-19 suffering from permanent kidney injury. For example, preclinical studies using iPSC-derived renal precursor cells have shown the ability for these cells to engraft into damaged tubules and improve renal function (124). In addition, Swaminathan et al. (125) conducted a phase 2 trial using intra-aortic allogenic MSCs in the setting of postcardiac surgeryrelated AKI. However, the results showed no significant improvement in time to recover from AKI, dialysis use, or 30-day mortality. A phase 1 clinical trial, which is not yet recruiting, is aiming to assess the efficacy and safety of allogeneic MSCs infused via continuous renal replacement therapy (CRRT) in patients with COVID-19 with AKI undergoing CRRT (NCT04445220). Patients will be divided into three arms: low dose (2.5 107 cells), high dose (7.5 107 cells), and control. These studies could shed light on a possible role for cell therapies for the treatment of COVID-related renal damage.

Hematological and vascular sequela, especially hypercoagulability and disseminated intravascular coagulation (DIC), are serious manifestations of SARS-CoV-2 (126). The hypercoagulable state increases the risk of venous thromboembolism, which can lead to ischemic stroke and multisystem organ failure via microemboli (127). Rates of venous thromboembolism in critically ill patients with COVID-19 have been estimated to be as high as 31% (128). Moreover, Tang et al. (129) reported that 70% of deceased patients met criteria for DIC. The hypercoagulable state may be related to stimulated production of antiphospholipid antibodies and complement activation, vascular endothelial damage, and prolonged immobility in the ICU (130). Although the hypercoagulable state is likely due to a variety of factors, endothelial disruption is one potential cause that may contribute to multisystem end-organ damage in COVID-19 (131). CD34+ cells, hematopoietic stem cells that can give rise to endothelial progenitors and restore vasculature, have been approved for an investigational new drug by the FDA to assess their efficacy and safety for lung damage repair. CD34+ cells are thought to promote vascular regeneration to counter ischemic damage and have shown efficacy and safety in trials evaluating their potential in cardiac and critical limb ischemia (132). Cord blood CD34+ cells also showed protective effects on acute lung injury induced by lipopolysaccharide challenge in mice, similar to another study that showed that peripheral blood CD34+ cells attenuated acute lung injury induced by oleic acid in rats (133, 134). Hence, therapy with CD34+ cells could prove feasible for promoting vascular growth in the lungs of patients with COVID-19 suffering from significant pulmonary damage (NCT04522817).

Overall, cell therapies show great promise in several diseases, and data from other studies suggest that certain cell therapies may be applicable in particular pathogenesis aspects of COVID-19. Specific factors such as dosing of the cells, route of administration, allogenic versus autologous cells, role of immunosuppressive therapy, tolerance of treatment in elderly patients, role of extracellular vesicles, and readouts of effectiveness need to be better delineated. As an example, risk for severe illness with COVID-19 increases with age. There are lessons to be learned about recipient age from studies using hematopoietic stem cell transplantation (HSCT) or MSCs. For instance, HSCT studies have shown that patient age is correlated with transplant-related morbidity and mortality, but improvements such as the use of cytokines and less toxic or reduced conditioning have allowed older patients to receive these therapies. In the context of MSCs, a study conducted to evaluate patient age on the efficacy of MSC cell therapy in ischemic cardiomyopathy showed that older patients did not have an impaired response. Although these studies are not directly translatable to other cell types or patients with COVID-19, they nevertheless represent a starting point for future investigation (135140). Cell dosing and number of injections should be tailored to patient-specific responses and tolerance of treatment. Route of administration should be localized as much as possible to reduce the risk of unintentional side effects in distant organs while maximizing efficacy at the infected organ system. Disseminated coronavirus involving multiple organ systems, for example, may benefit from intravenous infusion of cell therapy to allow treatment to reach multiple infected organs. Various routes of administration have been previously explored for respiratory and pulmonary diseases including intravenous, intratracheal instillation, inhalation, aerosolization, and nebulizers. Intratracheal instillation could be advantageous, as it provides highly precise, local delivery to the respiratory tract using a small dose; however, instillation is highly nonphysiological and may result in inconsistent and heterogeneous deposition focused on the upper airways (141). Five clinical trials for lung cell therapies have used aerosolization as the route of administration (NCT04313647, NCT04473170, NCT04389385, NCT04491240, and NCT04276987). This route of administration may be preferred because of the potentially broader distribution of cells in the lung while reducing the probability of cell damage and loss (141).

Another interesting avenue to consider is the use of a combination of various cell therapies. MSCs, for example, have been studied for their synergistic effects with other cell types, including pulmonary endothelial cells and epithelial cells (142). For instance, MSCs were shown to stimulate endothelial progenitors in patients with heart failure and preserve endothelial integrity after hemorrhagic shock (143, 144). These findings could support investigation of both cell types as a combination cell therapy.

From a scalability standpoint, allogenic or off-the-shelfbased therapies that are either human leukocyte antigen (HLA)matched or do not have HLAs present would be favored over autologous cells. HLA matching or depletion may also reduce the need for immunosuppression. Clinical trial readouts should include COVID-19related outcomes and organ function related to the cell therapy being administered. Last, the idea of leveraging the field of synthetic biology to further adapt engineered cell lines should also be considered. For example, cell therapies that modulate expression of viral entry proteins, decrease residual potentially tumorigenic pluripotent cells, or adopt genome-scale mammalian translational recoding to confer viral resistance could be of keen advantage (145, 146).

B. Diao, C. Wang, R. Wang, Z. Feng, J. Zhang, H. Yang, Y. Tan, H. Wang, C. Wang, L. Liu, Y. Liu, Y. Liu, G. Wang, Z. Yuan, X. Hou, L. Ren, Y. Wu, Y. Chen, Human kidney is a target for novel severe acute respiratory syndrome coronavirus 2 infection. 12, 2506 (2020).

Read more:
Cell therapy strategies for COVID-19: Current approaches and potential applications - Science Advances

Posted in Stem Cells | Comments Off on Cell therapy strategies for COVID-19: Current approaches and potential applications – Science Advances

Diabetic patient receives stem cell therapy – The Hindu

Posted: August 18, 2021 at 2:00 am

In a pioneering off-the-shelf drug treatment in place of a surgical procedure, Maruti Hospital on Tuesday inaugurated its Regenerative Medicine Department by administering stem cell therapy for a diabetic patient who lost four toes on both feet.

Developed by pharma major, Cipla, in collaboration with Bengaluru-based, Stempeutics Research, over 14 years, it is available (on order) in vials of 150 million and 200 million cells harvested from healthy individuals and costs between 1.5 to 2 lakh.

Diabetic foot ulcers/critical limb ischemia prevents the leg and feet from receiving adequate oxygen and nutrients needed for proper function. The stell cells are injected into the affected leg to promote new blood vessels growth called angiogenesis. It helps avoid amputation if given before gangrene sets in.

Stem cell treatment will help to improve blood circulation in the feet of the patient. The new method allows patients from any place to access this treatment in a ready-to-use procedure. We hope to use customised variations of this therapy for people with other medical ailments in the future, and reduce the dependence on transplants, said V. R. Ravi, orthopaedic surgeon, Maruti Hospital, said addressing the media.

The drug was produced in a carefully monitored processes, with mesenchymal stromal cell derived from the bone marrow of healthy donors in the age group of 20-25 years. It was transported from Bengaluru to Tiruchi by car with liquid nitrogen packs to keep the drug chilled. It was brought to room temperature within four hours and used on the patient.

B.N.Manohar, Managing Director and Chief Executive Officer, Stempeutics, was also present.

See the original post:
Diabetic patient receives stem cell therapy - The Hindu

Posted in Stem Cells | Comments Off on Diabetic patient receives stem cell therapy – The Hindu

New Stem cell conveying hydrogel could assist the heart with recuperating myocardial ischemia – Microbioz India

Posted: August 18, 2021 at 2:00 am

When blood vessels that feed the heart become blocked, damage to the heart muscle can occur and this can affect cardiac function. By stimulating the formation of new blood vessels, a new stem-cell-carrying gel helps mice overcome this condition called myocardial Infarction. The stem cell delivery system was developed by scientists from Kansai University, Japan. It is published in Science and Technology of Advanced Materials.

The hydrogel acts like a scaffold to hold the stem cells in place at injection site and keep them alive longer. The stem cells release cytokines, which stimulate the formation blood vessels and help the heart to recover. The gel is biodegradable so that it eventually dissolves and can be discarded by the body. Image credit: Kansai University

In their application, the team used stem cells from fat tissue. These stem cells, also known as adipose derived stem cells, have been used in the treatment of damaged cardiac tissue due to reduced blood flow. This is called myocardial Ischemia. Once injected into damaged tissue, the stem cells are supposed to release stimulants that can help regenerate blood vessels. However, they are not able to be retained in the tissue or survive long enough. Scientists have also found that injecting biodegradable hydrogels, which are cell-free, into damaged heart tissue can help partial recovery.

They first created hydrogels that could hold stem cells in place longer at the site where there is tissue damage. They are best used at room temperature. This allows you to easily mix the stem cells. The solution reacts with the body to heat and transforms into a gel when it is injected into the organ.

One hydrogel was particularly good at keeping its gel state. It was made from a mixture of molecules called tri-PCG with acrolyl group attached. The tri-PCG-acryl mixture was then combined with a polythiol derivative.

The team also added stem cells from adipose tissue to the hydrogel. They then observed how long they lived in petri dishes as well as the production of different genes and substances.

The stem cells were able to survive in our injectable hydrogel and released molecules that stimulate blood vessel formation, improving heart function and making it effective for treatment of ischemic heart.

Yuichi Ohya, Bioengineer, Kansai University

After confirming safety, the team plans to next test the therapy on larger animals and then conduct clinical trials in humans. They plan to use their injectable hydrogel for immune cells to treat cancer and in vaccines against viral infections.

Source:

Journal reference:

Yoshizaki, Y.,et al.(2021) Cellular therapy for myocardial ischemia using a temperature-responsive biodegradable injectable polymer system with adipose-derived stem cells.Science and Technology of Advanced Materials.doi.org/10.1080/14686996.2021.1938212.

Go here to see the original:
New Stem cell conveying hydrogel could assist the heart with recuperating myocardial ischemia - Microbioz India

Posted in Stem Cells | Comments Off on New Stem cell conveying hydrogel could assist the heart with recuperating myocardial ischemia – Microbioz India

Animal Stem Cell Therapy Market Technological Growth 2021-2026 with Types, Applications and Top Companies – The Market Writeuo – The Market Writeuo

Posted: August 18, 2021 at 2:00 am

The market study on the global Animal Stem Cell Therapy market will encompass the entire ecosystem of the industry, covering major regions namely North America, Europe, Asia Pacific, South America, Middle East & Africa, and the major countries falling under those regions.

This report includes the estimation of market size for value (million USD) and volume (K Units). Both top-down and bottom-up approaches have been used to estimate and validate the market size of Global Animal Stem Cell Therapy market, to estimate the size of various other dependent submarkets in the overall market. Key players in the market have been identified through secondary research, and their market shares have been determined through primary and secondary research. All percentage shares, splits, and breakdowns have been determined using secondary sources and verified primary sources.

The market study covers the Animal Stem Cell Therapy market size across different segments. It aims at estimating the market size and the growth potential across different segments, including application, type, organization size, vertical, and region. The study further includes an in-depth competitive analysis of the leading market players, along with their company profiles, key observations related to product and business offerings, recent developments, and market strategies.

Download Sample PDF at https://www.insidemarketreports.com/sample-request/14/873553/Animal-Stem-Cell-Therapy

Leading players of the Animal Stem Cell Therapy Market covered in this report are Medivet Biologics LLC, VETSTEM BIOPHARMA, J-ARM, U.S. Stem Cell, Inc, VetCell Therapeutics, Celavet Inc., Magellan Stem Cells, Kintaro Cells Power, Animal Stem Care, Animal Cell Therapies, Cell Therapy Sciences, Animacel,

The report is segmented based on product type are Dogs, Horses, Others, etc.

Major applications of the Animal Stem Cell Therapy market is segmented as Veterinary Hospitals, Research Organizations, etc.

Animal Stem Cell Therapy Market Regional Segment Analysis includes Regional Consumption Volume, Revenue and Growth Rate 2016-2026. Countries covered in this report are United States, Canada, Mexico, Brazil, Argentina, Columbia, Chile, Peru, Germany, UK, France, Italy, Russia, Spain, Netherlands, Turkey, Switzerland, GCC, North Africa, South Africa, China, Southeast Asia, India, Japan, Korea, Western Asia.

An Overview of the Impact of COVID-19 on this Market:

Effect of COVID-19: Animal Stem Cell Therapy Market report investigate the effect of Coronavirus (COVID-19) on the Animal Stem Cell Therapy industry. Since December 2019, the COVID-19 infection spread to practically 180+ nations around the world with the World Health Organization pronouncing it a general wellbeing crisis. The worldwide effects of the Covid infection 2020 (COVID-19) are now beginning to be felt, and will essentially influence the Animal Stem Cell Therapy market in 2020 and 2021.

Notwithstanding, this also will pass. Rising help from governments and a few organizations can help in the battle against this exceptionally infectious illness. There are a few ventures that are battling and some are flourishing. Generally speaking, pretty much every area is expected to be affected by the pandemic.

We are taking persistent endeavours to assist your business with maintaining and develop during COVID-19 pandemics. In view of our experience and aptitude, we will offer you an effective examination of Covid flare-up across enterprises to assist you with setting up whats to come.

Cautious assessment of the components molding the Animal Stem Cell Therapy market size, share, and the development direction of the market;

Get the insights to understand the Impact of COVID19 and drive the business strategies: https://www.insidemarketreports.com/covid-19/14/873553/Animal-Stem-Cell-Therapy

This study will address some of the most critical questions which are listed below:

Major Points from the Table of Contents

1 Animal Stem Cell Therapy Market Overview

2 Global Animal Stem Cell Therapy Market Competition by Manufacturers

3 Global Animal Stem Cell Therapy Capacity, Production, Revenue (Value) by Region)

4 Global Animal Stem Cell Therapy Supply (Production), Consumption, Export, Import by Region

5 Global Animal Stem Cell Therapy Production, Revenue (Value), Price Trend by Type

6 Global Animal Stem Cell Therapy Market Analysis by Application

7 Global Animal Stem Cell Therapy Manufacturers Profiles/Analysis

8 Animal Stem Cell Therapy Manufacturing Cost Analysis

9 Industrial Chain, Sourcing Strategy and Downstream Buyers

10 Marketing Strategy Analysis, Distributors/Traders

11 Market Effect Factors Analysis

12 Global Animal Stem Cell Therapy Market Forecast

13 Research Findings and Conclusion

14 Appendix

Why Inside Market Reports:

For all your Research needs, reach out to us at:

Email: [emailprotected]

Phone: +1-617-230-0741

Mobile Payment Technology Market and Ecosystem by Production, Prospects, Consumption, Cost Structure, Competitive Landscape

Turbine Gearbox for Thermal Power Market Technological Growth 2021-2026 with Types, Applications and Top Companies

Orthopedic Surgical Navigation Systems Market: Classification, Opportunities, Types, Applications, Status and Forecast to 2026

See the original post here:
Animal Stem Cell Therapy Market Technological Growth 2021-2026 with Types, Applications and Top Companies - The Market Writeuo - The Market Writeuo

Posted in Stem Cells | Comments Off on Animal Stem Cell Therapy Market Technological Growth 2021-2026 with Types, Applications and Top Companies – The Market Writeuo – The Market Writeuo

Stem Cell Therapy Market worth $40.3 billion by 2027 Exclusive Report by CoherentMarketInsights – PharmiWeb.com

Posted: August 18, 2021 at 2:00 am

The Stem Cell Therapy Market report provides a quick description about market status, size, companies share, growth, opportunities and upcoming trends. This report includes the corporate profile, values that the challenges and drivers & restraints that have a serious impact on the industry analysis. The information within the report that help form the longer term projections during the forecast year. The up so far analysis to assists in understanding of the changing competitive analysis. Additionally, the market strategies including moderate growth during the years.

The research on Stem Cell Therapy market scenario which will affect the overview the forecast period, including as opportunities, prime challenges, and current/future trends. To supply an in-depth analysis of all Stem Cell Therapy regions included within the report into sections to supply a comprehensive competitive analysis.

Get Your Sample Copy of the Stem Cell Therapy Market Report 2021

Some of the leading manufacturers and suppliers of the Stem Cell Therapy market are Magellan, Medipost Co., Ltd, Osiris Therapeutics, Inc., Kolon TissueGene, Inc., JCR Pharmaceuticals Co., Ltd., Anterogen Co. Ltd., Pharmicell Co., Inc., and Stemedica Cell Technologies, Inc.

Stem cells are divided into two major classes; pluripotent and multipotent. Pluripotent stem cells are replicating cells, which are derived from the embryo or fetal tissues. The pluripotent stem cells facilitate the development of cells and tissues in three primary germ layers such as mesoderm, ectoderm, and endoderm.

Market Dynamics

Increasing expansion of facilities by market players for stem cell therapies is expected to propel growth of the stem cell therapy market over the forecast period. For instance, in January 2018, the University of Florida, U.S. launched the Center for Regenerative Medicine that is focused on development of stem cell therapies for the treatment of damaged tissue and organ. The Centre for Regenerative Medicines is divided into two segments such as focus groups and shared services. Focus groups such as research and development activities for stem cell therapies; and the shared services segment offers technical resources related to stem cell therapies.

Furthermore, rising collaboration activities by key players are expected to drive growth of the global stem cell therapy market. For instance, in May 2018, Procella Therapeutics and Smartwise, a medtech company entered into a collaboration with AstraZeneca Pharmaceuticals. Under this collaboration, AstraZeneca utilized Procella Therapeutics stem cell technology for the development of stem cell therapies in cardiovascular diseases. Moreover, in April, 2019, CelluGen Biotech and FamiCord Group collaborated to develop new stem cell-based drugs and advanced medical therapies (ATMP)

What Stem Cell Therapy Market Research Report Covers?

This report covers definition, development, market status, geographical analysis of Stem Cell Therapy market.

Competitor analysis including all the key parameters of Stem Cell Therapy market

Market estimates for at least 7 years

Market Trends (Drivers, Constraints, Opportunities, Threats, Challenges, Investment Opportunities, and proposals)

Strategic proposals in key business portions dependent available estimations

Company profiling with point by point systems, financials, and ongoing improvements

Mapping of the most recent innovative headways and Supply chain patterns

Buy This Complete A Business Report With Flat US $2000 Off @https://www.coherentmarketinsights.com/promo/buynow/2848

Increasing application of stem cells for the treatment of patients with blood-related cancers, spinal cord injury and other diseases are the leading factors that are expected to drive growth of stem cell therapy market over the forecast period. According to the National Spinal Cord Injury Statistical Center, 2016, the annual incidence of spinal cord injury (SCI) is approximately 54 cases per million population in the U.S. or approximately 17,000 new SCI cases each year.

Moreover, according to the Leukemia and Lymphoma Society, 2017, around 172,910 people in the U.S. were diagnosed with leukemia, lymphoma or myeloma in 2017, thus leading to increasing adoption of stem cells for its efficient treatment. Increasing product launches by key players such as medium for developing embryonic stem cells is expected to propel the market growth over the forecast period.

For instance, in January 2019, STEMCELL Technologies launched mTeSR Plus, a feeder-free human pluripotent stem cell (hPSC) maintenance medium for avoiding conditions associated with DNA damage, genomic instability, and growth arrest in hPSCs. With the launch of mTeSR, the company has expanded its portfolio of mediums for maintenance of human embryonic stem (ES) cells and induced pluripotent stem (iPS) cells. Increasing research and development of induced pluripotent stem cells coupled with clinical trials is expected to boost growth of the stem cell therapy market over the forecast period.

For instance, in April 2019, Fate Therapeutics in collaboration with UC San Diego researchers launched Off-the-shelf immunotherapy (FT500) developed from human induced pluripotent stem cells. The therapy is currently undergoing clinical trials for the treatment of advanced solid tumors.

Ask for PDF sample copy of the Stem Cell Therapy market report

Main points in Stem Cell Therapy Market Report Table of Content

Chapter 1 Industry Overview

1.1 Definition

1.2 Assumptions

1.3 Research Scope

1.4 Market Analysis by Regions

1.5 Global Stem Cell Therapy Market Size Analysis from 2021 to 2027

11.6 COVID-19 Outbreak: Stem Cell Therapy Industry Impact

Chapter 2 Global Stem Cell Therapy Competition by Types, Applications, and Top Regions and Countries

2.1 Global Stem Cell Therapy (Volume and Value) by Type

2.3 Global Stem Cell Therapy (Volume and Value) by Regions

Chapter 3 Production Market Analysis

3.1 Global Production Market Analysis

3.2 Regional Production Market Analysis

Chapter 4 Global Stem Cell Therapy Sales, Consumption, Export, Import by Regions (2016-2021)

Chapter 5 North America Stem Cell Therapy Market Analysis

Chapter 6 East Asia Stem Cell Therapy Market Analysis

Chapter 7 Europe Stem Cell Therapy Market Analysis

Chapter 8 South Asia Stem Cell Therapy Market Analysis

Chapter 9 Southeast Asia Stem Cell Therapy Market Analysis

Chapter 10 Middle East Stem Cell Therapy Market Analysis

Chapter 11 Africa Stem Cell Therapy Market Analysis

Chapter 12 Oceania Stem Cell Therapy Market Analysis

Chapter 13 South America Stem Cell Therapy Market Analysis

Chapter 14 Company Profiles and Key Figures in Stem Cell Therapy Business

Chapter 15 Global Stem Cell Therapy Market Forecast (2021-2027)

Chapter 16 Conclusions

View Press Release For More Information

Related Reports:

Stem Cell Assay Market, By Product Type (Instruments and Reagents & Kits), By Cell Type (Adult Stem Cells and Human Embryonic Stem Cells), By Process (Cell Culture, Engineering, Differentiation, Characterization, and Others), By Application (Research, Drug Discovery & Development, and Regenerative Medicine), and By Region Global Industry Insights, Trends, Outlook, and Opportunity Analysis, 2020-2027

Stem Cell Banking Market, by Source Type (Cord Blood and Cord Tissue), by Service Type (Collection and Transportation, Processing, Analysis and Storage), by Application (Leukemia, Diabetes, Lymphoma, Cerebral Palsy, Thalassemia and Others), and by Region Global Trends, and Forecast to 2027

Cell Culture Media for Vaccine Market by Media Type (Bovine Derived, Porcine Derived, and Animal Free/Serum Free Cell Culture), by Application (Human Vaccine and Animal Vaccine), and by Region (North America, Latin America, Europe, Asia Pacific, Middle East, and Africa) Size, Share, Outlook, and Opportunity Analysis, 2020-2027

About Us:

Coherent Market Insights is a global market intelligence and consulting organization focused on assisting our plethora of clients to achieve transformational growth by helping them make critical business decisions. We are headquartered in India, having an office at global financial capital in the U.S. and sales consultants in the United Kingdom and Japan. Our client base includes players from across various business verticals in over 150 countries worldwide. We pride ourselves in catering to clients across the length and width of the horizon, from Fortune 500 enlisted companies to not-for-profit organizations, and startups looking to establish a foothold in the market. We excel in offering unmatched actionable market intelligence across various industry verticals, including chemicals and materials, healthcare, and food & beverages, consumer goods, packaging, semiconductors, software and services, Telecom, and Automotive. We offer syndicated market intelligence reports, customized research solutions, and consulting services.

Contact:

Coherent Market Insights1001 4th Ave, #3200 Seattle, WA 98154, U.S.Email:sales@coherentmarketinsights.comUnited States of America: +1-206-701-6702United Kingdom: +44-020-8133-4027Japan: +050-5539-1737India: +91-848-285-0837

Read more:
Stem Cell Therapy Market worth $40.3 billion by 2027 Exclusive Report by CoherentMarketInsights - PharmiWeb.com

Posted in Stem Cells | Comments Off on Stem Cell Therapy Market worth $40.3 billion by 2027 Exclusive Report by CoherentMarketInsights – PharmiWeb.com

Atf3 and Rab7b genes drive regeneration in mature cells – Baylor College of Medicine News

Posted: August 18, 2021 at 2:00 am

When an injury occurs, damaged cells need to be replaced. Stem cells, known as the go-to cells when new specialized cells need to be produced, are rare in adult tissues, so the job often falls to differentiated, or mature, cells.

Dr. Jason Mills and his lab have been working on identifying the genes driving mature cells to return to a regenerative state, a process called paligenosis.

My lab has been promoting the idea that given that cells in all organs use similar functions like mitosis and apoptosis, theres likely to be a conserved genetic program for how mature cells become regenerative cells, said Mills, senior author of the study and professor of medicine gastroenterology,pathology and immunologyandmolecular and cellular biologyat Baylor. The research was conducted while his lab was atWashington University School of Medicine in St. Louis.

To begin paligenosis and reenter the cell cycle, mature cells must first go through the process of autodegredation, breaking down larger structures used in specialized cell function. Mills and his team, led by first author Dr. Megan Radyk, a postdoctoral associate at the Washington University School of Medicine in St. Louis at the time of research, found that the genes Atf3 and Rab7b are upregulated in gastric and pancreatic digestive-enzyme-secreting cells of mice during autodegredation, and return to normal expression before mitosis.

The researchers showed that Atf3 activates Rab7b, which directs lysosomes to begin dismantling cell parts not needed for regeneration. But when Atf3 was not present, Rab7b did not trigger autodegredation.

The team also found Atf3 and Rab7b expression were consistent in paligenosis across other organs and organisms. Similar gene expression also appeared in precancerous gastric lesions in humans. According to Mills, the discoveries in this research are foundational to understanding how repetitive injury and paligenosis may impact cancer.

The more tissue damage you have, the more youre calling mature cells back into regeneration duty, said Mills, co-director of theTexas Medical Center Digestive Disease Center. Theres emerging evidence that, when these cells go through paligenosis, they dont check for DNA damage well. The cells are storing DNA mutations when they return to their differentiated function. Over time, they become so damaged that they cant go back to normal function and instead keep replicating.

Its our belief that paligenosis is at the heart of cancer development.

This research also provides groundwork for potential therapeutic targets. Existing drugs like hydroxychloroquine can be used to inhibit autodegredation, therefore stopping paligenosis.

According to Mills, further study is required to determine whether drugs targeting autodegredation can be used in conjunction with cancer treatments to stop cells from replicating.

The complete study is published in EMBO Reports.

For a full list of authors, their contributions to this work and sources of support, see the publication.

By Molly Chiu

Read more:
Atf3 and Rab7b genes drive regeneration in mature cells - Baylor College of Medicine News

Posted in Stem Cells | Comments Off on Atf3 and Rab7b genes drive regeneration in mature cells – Baylor College of Medicine News

Page 674«..1020..673674675676..680690..»