Page 7«..6789..2030..»

Category Archives: Stem Cells

Treatment Choices for AML Were Increasingly Influenced by Immunological Responses – Physician’s Weekly

Posted: September 8, 2022 at 1:56 am

One of the most frequently altered genes in AML is nucleophosmin1 (NPM1), which is frequently linked to a good prognosis. Immunological responses increasingly influenced treatment choices for AML. However, it was unclear how immune checkpoint inhibition functions. For a study, researchers sought to determine the particular immune responses of AML patients to NPM1, PRAME, Wilms tumor 1, RHAMM, and 3 additional leukemia-associated antigens (LAA).

Using colony-forming immunoassays and flow cytometry, they examined T cell responses against leukemic progenitor/stem cells (LPC/LSC). In addition, comparing cells from NPM1 mutant and NPM1 wild-type AML patients, they investigated whether immune checkpoint suppression with the anti-programmed death 1 antibody improved the immune response to stem cell-like cells.

Nivolumab, an anti-PD-1 antibody, was reported to enhance the number of LAA-stimulated cytotoxic T cells and to have a cytotoxic impact on LPC/LSC. When the immunogenic epitope was obtained from the area of NPM1 that had been altered, the impact was best against NPM1mut cells and the effects were strengthened by the addition of anti-PD-1.

The results implied that the immune checkpoint inhibitor anti-PD-1 may be used to treat individuals with NPM1 mutant AML and that this treatment, in combination with NPM1-mutation specific directed immunotherapy, may be even more successful for this particular subset of patients.

Reference: onlinelibrary.wiley.com/doi/10.1111/bjh.18326

Read more here:
Treatment Choices for AML Were Increasingly Influenced by Immunological Responses - Physician's Weekly

Posted in Stem Cells | Comments Off on Treatment Choices for AML Were Increasingly Influenced by Immunological Responses – Physician’s Weekly

BioRestorative Therapies to Present at the H.C. Wainwright 24th Annual Global Investment Conference on September 14 – StreetInsider.com

Posted: September 8, 2022 at 1:56 am

Get inside Wall Street with StreetInsider Premium. Claim your 1-week free trial here.

BioRestorative Therapies to highlight addition of Phase 2 clinical trial for chronic lumbar disc disease to its mid-stage pipeline.

Melville, NY., Sept. 07, 2022 (GLOBE NEWSWIRE) -- BioRestorative Therapies. ("BioRestorative" or the "Company") (Nasdaq:BRTX), a clinical stage company focused on the development of first-in-class cell-based therapies,today announced that it will present on September 14, 2022 at the H.C. Wainwright 24th Annual Global Investment Conference being held virtuallySeptember 12-14, 2022.

Lance Alstodt, President and CEO of the Company,will be speaking and providing an update onthe Company's expanded pipeline and near-term catalysts of itsongoing Phase 2 clinical trial targeting chronic lumbar disc disease.

Further information on this conference, including registration links, can be found below:

H.C. Wainwrights 24th Annual Global Investment ConferenceSeptember 12-14, 2022@ Lotte New York Palace Hotel - New York, NYPresentation Information: BioRestorative on-demand presentation will be livestarting on Wednesday, September 14, 2022 at 11:30am EDTPresenter: Lance Alstodt, CEO & President, BioRestorative Therapies.Registration Link:www.hcwevents.com/annualconferenceWebcasting Link:https://journey.ct.events/view/8518bd6c-814c-477d-a15f-973612fc9e56

About BioRestorative Therapies, Inc.

BioRestorative Therapies, Inc. (www.biorestorative.com) develops therapeutic products using cell and tissue protocols, primarily involving adult stem cells. Our two core programs, as described below, relate to the treatment of disc/spine disease and metabolic disorders:

Disc/Spine Program (brtxDISC): Our lead cell therapy candidate, BRTX-100, is a product formulated from autologous (or a persons own) cultured mesenchymal stem cells collected from the patients bone marrow. We intend that the product will be used for the non-surgical treatment of painful lumbosacral disc disorders or as a complementary therapeutic to a surgical procedure. The BRTX-100 production process utilizes proprietary technology and involves collecting a patients bone marrow, isolating and culturing stem cells from the bone marrow and cryopreserving the cells. In an outpatient procedure, BRTX-100 is to be injected by a physician into the patients damaged disc. The treatment is intended for patients whose pain has not been alleviated by non-invasive procedures and who potentially face the prospect of surgery. Pursuant to authorization received from the Food and Drug Administration, we have commenced a Phase 2 clinical trial using BRTX-100 to treat chronic lower back pain arising from degenerative disc disease.

Metabolic Program (ThermoStem): We are developing a cell-based therapy candidate to target obesity and metabolic disorders using brown adipose (fat) derived stem cells to generate brown adipose tissue (BAT). BAT is intended to mimic naturally occurring brown adipose depots that regulate metabolic homeostasis in humans. Initial preclinical research indicates that increased amounts of brown fat in animals may be responsible for additional caloric burning as well as reduced glucose and lipid levels. Researchers have found that people with higher levels of brown fat may have a reduced risk for obesity and diabetes.

Forward-Looking Statements

This press release contains "forward-looking statements" within the meaning of Section 27A of the Securities Act of 1933, as amended, and Section 21E of the Securities Exchange Act of 1934, as amended, and such forward-looking statements are made pursuant to the safe harbor provisions of the Private Securities Litigation Reform Act of 1995. You are cautioned that such statements are subject to a multitude of risks and uncertainties that could cause future circumstances, events or results to differ materially from those projected in the forward-looking statements as a result of various factors and other risks, including, without limitation, those set forth in the Company's latest Form 10-K filed with the Securities and Exchange Commission and other public filings. You should consider these factors in evaluating the forward-looking statements included herein, and not place undue reliance on such statements. The forward-looking statements in this release are made as of the date hereof and the Company undertakes no obligation to update such statements.

CONTACT:Email: [emailprotected]

Read more from the original source:
BioRestorative Therapies to Present at the H.C. Wainwright 24th Annual Global Investment Conference on September 14 - StreetInsider.com

Posted in Stem Cells | Comments Off on BioRestorative Therapies to Present at the H.C. Wainwright 24th Annual Global Investment Conference on September 14 – StreetInsider.com

Global Recombinant Cell Culture Supplements Market Report 2022: Increasing Need for Immunotherapies and Stem Cell and Regenerative Medicines Presents…

Posted: September 8, 2022 at 1:56 am

Company Logo

Global Recombinant Cell Culture Supplements Market

Global Recombinant Cell Culture Supplements Market

Dublin, Sept. 05, 2022 (GLOBE NEWSWIRE) -- The "Global Recombinant Cell Culture Supplements Market: Focus on Pricing Analysis, Product, Application, Expression System, and Region - Analysis and Forecast, 2022-2032" report has been added to ResearchAndMarkets.com's offering.

The global recombinant cell culture supplements market was estimated at $308.6 million in 2021 and is expected to reach $1,188.6 million by 2032, growing at a CAGR value of 12.24% during the forecast period 2022-2032. The growth in the global recombinant cell culture supplements market is expected to be driven by the rising demand for cell culture supplements, increasing investment in life sciences research and development, as well as growing advantages of recombinant supplements over traditional animal-derived supplements.

Market Lifecycle Stage

The global recombinant cell culture market is increasing at a rapid pace. The growing need for animal-free supplements in cell culture applications is aiding the growth of the recombinant cell culture supplements market. Recombinant cell culture supplements play a crucial role in enhancing cell viability, maintaining a healthy culture, and customizing the cell culture in accordance with the needs of the individual.

Increasing demand for immunotherapy and stem cell and regenerative medicine research is one of the major opportunities in the recombinant cell culture supplements market. Several cell culture companies, and biopharmaceutical companies are working collaboratively on drug development and using recombinant cell culture supplements as a therapeutic means for applications in biological drugs. Furthermore, the market witnessed major mergers and acquisitions in the past four years. For instance, recently, in March 2022, Thermo Fisher Scientific Inc. acquired PeproTech, Inc., a company that specializes in the development and manufacturing of recombinant proteins, in a deal of $1.85 billion.

Story continues

Impact

Many biopharmaceutical products are being developed by utilizing the cell culture technique. The study of cell physiology and biochemistry is made possible through laboratory cell culture, which also opens up research avenues that are challenging to pursue in vivo. Controlling variables such as the culture media, culture conditions, population density, and growth rate makes it simple to assess the effects of medications or other substances on cultured cells.

Additionally, it allows analyzing the function of various genes and offers enormous potential in the field of genetics. It enables the assessment of harmful and carcinogenic chemicals on cells in the fields of oncology and virology and the comprehension of how different medications, viruses, and physical or chemical carcinogens interact.

Furthermore, the recombinant cell culture has various applications, such as research on vaccines, stem cells, gene therapy, and genetic engineering, as well as the creation of protein therapies manufacturing of genetically edited proteins such as monoclonal antibodies, insulin, and hormones.

Market Dynamics

Market Drivers

Increasing Demand for Recombinant Cell Culture Supplements Due to their Advantages

Increased Funding and Investment in Research and Development for Biopharmaceutical Products in Life Sciences Sector

Growing Number of Mergers and Acquisitions for Expanding Recombinant Cell Culture Supplements Portfolio

Push from Regulatory Bodies to Utilize Animal-Free Media Supplements in Cell Culture Process

Market Restraints

Market Opportunities

Market Segmentationby Product

Recombinant Albumin (rAlbumin)

Recombinant Insulin (rInsulin)

Recombinant Epidermal Growth Factor (rEGF)

Recombinant Interleukin Growth Factor (rILGF)

Recombinant Transferrin (rTransferrin)

Recombinant Trypsin (rTrypsin)

Recombinant Insulin-like Growth Factor (rIGF)

Recombinant Stem Cell Factor (rSCF)

Recombinant Aprotinin (rAprotinin)

Recombinant Lysozyme (rLysozyme)

Others

by Application

by Expression System

Mammalian

E. Coli

Yeast

Others

by Region

North America

Europe

Asia-Pacific

Latin America

Rest-of-the-World

Demand - Drivers and LimitationsFollowing are the demand drivers for the global recombinant cell culture supplements market:

Increasing Demand for Recombinant Cell Culture Supplements Due to their Advantages

Increased Funding and Investment in Research and Development for Biopharmaceutical Products in Life Sciences Sector

Growing Number of Mergers and Acquisitions for Expanding Recombinant Cell Culture Supplements Portfolio

Push from Regulatory Bodies to Utilize Animal-Free Media Supplements in Cell Culture Process

The market is expected to face some limitations too due to the following challenges:

Key Topics Covered:

1 Market Overview

2 Product, $ Mn, 2021-2032

3 Application, $Mn, 2021-2032

4 Expression System, $Mn, 2021-2032

5 Region, $Mn, 2021-2032

6 Competitive Landscape

7 Company Profiles

Companies Mentioned

Abcam plc.

BBI Solutions

Corning Incorporated

FUJIFILM Irvine Scientific, Inc.

Gemini Bioproducts, LLC

HiMedia Laboratories

InVitria

Kingfisher Biotech, Inc.

Lonza Group AG

Merck KGaA

Novus Biologicals, LLC

R&D Systems, Inc.

Sartorius AG

Sino Biological Inc.

STEMCELL Technologies Inc.

Thermo Fisher Scientific Inc.

For more information about this report visit https://www.researchandmarkets.com/r/2s1ob7

Attachment

See the article here:
Global Recombinant Cell Culture Supplements Market Report 2022: Increasing Need for Immunotherapies and Stem Cell and Regenerative Medicines Presents...

Posted in Stem Cells | Comments Off on Global Recombinant Cell Culture Supplements Market Report 2022: Increasing Need for Immunotherapies and Stem Cell and Regenerative Medicines Presents…

CAR-T Beyond CGTs In Development In 2022 – BioProcess Online

Posted: September 8, 2022 at 1:56 am

By Maria Aspioti and Paolo Siciliano, PA Consulting

The world of advanced therapeutics medicinal products (ATMPs) and, in particular, the cell and gene therapies (C>) space has been experiencing outstanding growth over the last few years, with a number of therapies transitioning from clinical research into regular clinical practice.In recent years, new cell types and new technologies have been used to overcome challenges posed by current treatments and by the nature of the targeted diseases, thus enabling us to treat, and in some cases potentially cure, severe disorders. The scientific and R&D efforts led to the discovery of new ways to engineer cells, enabling some of the most outstanding hurdles in complex disease areas such as oncology, cardiovascular, neurologic, and metabolic disorders to be addressed.

While these technology and scientific advancements are all positive and are promising signs of a growing and thriving sector, the other side of the coin shows a highly fragmented market, with very high levels of uncertainty on what type(s) of approaches will be successful in providing patients with a true alternative and which approaches will quickly become obsolete due to technical or commercial limitations. So, how do companies and investors in the CGT space hedge their bets in a fast-evolving and highly uncertain market?

In this article, we review the main cell technologies currently being developed in clinical research for oncology and other therapeutic areas, including examples of studies being conducted, developers, modes of action, and benefits of the different types of cell therapies, and share insight on how to avoid pitfalls and prepare for rapid market and technological directional changes.

Chimeric antigen receptor (CAR) T cells have been dominating the C> field for years, resulting in the approval and commercialization of Kymriah, the first therapy of this kind, in 2017.

Since then, CAR-T therapies have been the most researched type of cell therapy globally and five more products based on this technology have been approved for the treatment of various types of blood cancers worldwide (Yescarta, Abecma, Tecartus, Breyanzi, and Carvykti).

The current commercially available CAR T cell therapies (which are all autologous) have shown efficacy in the treatment of hematologic cancers such as acute lymphoblastic leukemia (ALL), chronic lymphocytic leukemia (CLL), non-Hodgkin's lymphoma (NHL), diffuse large B cell lymphoma (DLBCL), and other B cell malignancies. Given the increasing number of clinical studies utilizing CAR T cell therapies and the already successful application in cancer patients, this class of biotherapeutics is likely to dominate the C> market and R&D space for the next few years. This is also shown by the predicted CAGR of 30.6% over the period 2021-2031, which should lead to a total CAR T cell therapy market size of $23.2 billion in the next decade.

At present, there are 750 active CAR-T therapies in development across the globe (375 in clinical phases and 378 in preclinical stage). This represents over a 50% increase from 2019, when approximately 245 CAR-T therapies were in clinical development. Currently, CAR-T therapies still represent 31% of the clinical pipeline in C> (375 out of 1,191 active trials) The vast majority of these are in the early clinical development stage (predominantly Phase 1), with oncology counting for over 95% of the active CAR-T trials.

While still the predominant realm in C>, CAR-T therapies present some limitations.

Numerous allogeneic alternatives are being investigated to overcome some of the challenges faced by CAR-T therapies in oncology. In the field of adoptive cell immunotherapy for oncology, we are seeing an increasing exploitation of alternative cell sources with a high therapeutic potential that aim to evolve toward universal allogeneic alternatives to classic CAR-T therapies. Some examples include (further investigative product examples are shown in Table 1 [below]):

In addition to CAR-based technologies, the market is also seeing an increasing number of preclinical and clinical studies focusing on CAR-free cell therapy alternatives to cure cancer. The list of cell types is continuously growing, but we see five main categories that are showing promising results (investigative product examples are shown in Table 1):

Induced Pluripotent Stem Cells (iPSCs): iPSCs are a type of pluripotent stem cells that are generated ex vivo by treating nearly any human fully differentiated (somatic) cell (e.g., keratinocytes, fibroblasts, etc.) with the cocktail of small molecules described by the 2012 Nobel Laureate Shynia Yamanaka. iPSCs display several advantages over primary cells, including their virtually infinite proliferation capacity and amenability for genetic manipulation. Several T and NK cells derived from iPSC lines (iT and iNK, respectively) are currently under investigation, primarily for B cell lymphoma and advanced solid tumors.An example is represented by Shoreline Biosciences, which is developing a pipeline of iPSC-derived natural killer cell (iNK) and macrophage (iMACs) cellular immunotherapy candidates for the treatment of different types of cancers.

Mesenchymal Stem Cells (MSCs): MSCs are multipotent stem cells capable of self-renewal that are commonly found in the bone marrow but also in the umbilical cord, adipose tissue, and peripheral blood. In clinical trials, MSCs are used in cancer treatment either via direct transplantation (often used to support chemotherapy or radiotherapy), as genetically modified cell therapy, or as a carrier of anti-tumor agents like interferon , interleukins, bone morphogenic protein 4, and many others. At the end of 2021, 31 clinical trials concerning MSC-based therapies for cancer were registered on ClinicalTrials.gov. The majority of these studies focus on the direct infusion or transplantation of MSCs to treat cancer, while the remaining trials use engineered MSCs as vehicles of therapeutic agents such as cytokines or oncolytic viruses.MSCs are also being investigated for a wide range of non-oncology applications, including cardiovascular conditions as well as neurodegenerative disorders such as Alzheimers, multiple sclerosis, and amyotrophic lateral sclerosis. Brainstorm Cell Therapeutics is currently in the process of finalizing the regulatory filing for NurOwn (autologous MSC-NTF cells produced from autologous, bone marrow-derived mesenchymal stem cells) for the treatment of ALS.

Dendritic Cells: Dendritic cells (DCs) are antigen-presenting cells (APCs) that represent another valuable alternative to CAR-T therapies. In clinical settings, DCs find applications as vaccines owing to their ability to prepare the adaptive and innate immune system against specific tumors via presenting cancer-specific antigens. To date, the only FDA-approved DC-based vaccine is Provenge (sipuleucel-T, Dendreon), which targets patients with metastatic castration-resistant prostate cancer. Another DC-based medicinal product named Apceden, developed by Apac Biotech, was approved in India by the Central Drugs Standard Control organization in 2017. Currently, there are five Phase 1, 10 Phase 2, and five Phase 3 clinical trials ongoing that demonstrate the excitement around this cell therapy type.

Tumor Infiltrating Lymphocytes (TILs): TILs are immune cells that can infiltrate tumor masses and currently present an alternative therapeutic solution that has mainly been researched for the treatment of advanced solid tumor indications. Currently, TIL cell therapies are being explored at the clinical setting, predominantly for the treatment of melanoma with a number of other cancer indications also being under investigation.Two key players in this space are Achilles Therapeutics and Iovance Biotherapeutics, both of which are developing TIL-based therapies (Phase 2 trials) for the treatment of different types of cancer.

Regulatory T Cells (Tregs): Similar to TILs, biotherapeutics based on Tregs are also being investigated as a form of cell therapy for multiple indications. Treg-cell therapies are currently in early infancy with multiple opportunities being explored across a spectrum of indications such as type 1 diabetes, rheumatoid arthritis, multiple sclerosis, and others.Quell Therapeutics, GentiBio, and Sonoma (among others) are developing Treg-based therapies to address a number of autoimmune and alloimmune conditions.

Oncology is the field where the majority of C>s are commercially available currently. As described above, a number of biotherapeutics have been approved by the FDA and other regulatory agencies, including CAR T cell therapies as well as other immunotherapies such as talimogene laherparepvec (Imlygicby Amgen Inc.) and sipuleucel-T (Provenge by Dendreon Corp.).

While oncology has been driving R&D in C> since the beginning of the new wave of therapeutic innovation, the interest of academic groups, biotechnology firms, and large pharmaceutical companies in different disease areas for ATMPs is rapidly expanding. This is particularly the case for in vivo gene therapies, where we have recently seen the approval of products for the treatment of spinal muscular atrophy (Zolgensma by Novartis) and mutation-specific retinal dystrophy (Luxturna by Spark Therapeutics), as well as an increasing number of trials across a large spectrum of non-oncology applications, including several rare genetic disorders.

For cell therapies, in the last few years, a number of therapies have been launched on the market for non-oncology applications. These include Rethymic by Enzyvant Therapeutics (congenital athymia), Stratagraft (deep partial-thickness burns), Gintuit (epithelial damage), and Maci (cartilage damage). Several non-oncological biotherapeutics received approval for use in unrelated donor hematopoietic progenitor cell transplantation (Allocord, Clevecord, Ducord, Hemacord).

As indicated in our Cell & Gene Therapy 2040 Report, which looks at the future of the C> industry, the clinical development of C>s is predominantly aimed at cardiovascular, metabolic, neurological, inflammatory/autoimmune, and musculoskeletal disorders, with a particular focus on rare genetic conditions. Currently, oncology-unrelated Phase 3 trials focus on more than 20 different indications with over 25 lead companies involved.

Among the indications with the highest number of clinical studies, it is worth noting hemophilia A, for which Pfizer, Roche, and BioMarin Pharmaceutical developed similar C>s targeting coagulation factor VIII. BioMarin Pharmaceutical retains a slight commercial advantage as their asset also targets a different coagulation factor and is expected to reach approval by the end of 2022 both in Europe and the U.S. (approvals are expected for Pfizer in 2023 and Roche in 2024, both in the U.S. only).

Other indications that are seeing a surge in late-phase clinical trials include Duchenne muscular dystrophy and Crohns disease. For the former indication, Pfizer and Sarepta Therapeutics are currently recruiting patients for the virus-mediated administration of the gene encoding for microdystrophin to help rescue the muscle architecture. In Crohns disease, Takeda Pharmaceutical and Mesoblast Ltd. are the two front-runners. Notably, both are developing MSC-based therapies for this indication, with Takeda having already shown positive results (ClinicalTrials.gov Identifier: NCT03706456) and currently recruiting for two additional Phase 3 studies.

The field of C> is fast-growing and booming with novel technologies, new companies, and growing investment, with more and more positive results in treating, and even curing, life-threatening diseases. But how can organizations hedge their bets in such a fast-evolving and highly uncertain market?

Here are some tips on how different players in the CGT space can avoid pitfalls and better position themselves to succeed in this space:

Being aware of the C> landscape and how it is changing becomes paramount for developers. A clear view of the market and its evolution will enable developers to:

In addition, understanding the nature of the new biotherapeutics developed and how they are delivered is vital for C> manufacturers, healthcare professionals, and patients to enable a facilitated clinical application while reducing the overall costs of these transformative therapies.

Time to market is key to avoid a technology becoming obsolete in a fast-evolving market. The complexity of developing and launching new products in the C> market (these being therapies or technologies involved in their manufacturing) requires a level of investment, competencies, and capabilities that rarely are available in a single organization. Hence, innovators in this space should invest time and resources in identifying the right partners to support their product development, access the right technologies to manufacture their therapies, embrace digital tools early on to support the launch of their products, as well as work with experts to speed up the transition from R&D to clinical and commercial scale.

Focus is usually key in bringing new products and services to market in highly innovative sectors. However, to mitigate risk in a highly uncertain market, established pharma and biotech companies developing biotherapeutics should look at diversifying their portfolio through the development and/or acquisition of multiple C> platforms across different therapeutic areas. Similarly, equipment manufacturers should also look at how their current products and innovation portfolios can support the needs of different product lines, as certain technologies might quickly become obsolete if a specific set of therapies becomes predominant in the market.

Overall, the advent of C> therapies has the potential to revolutionize healthcare by providing therapies for rare, complex, and life-threatening diseases. Successful positioning of players in this flourishing market will require careful consideration of the evolving market dynamics coupled with successful go-to-market and risk mitigation strategies.

About The Authors:

Maria Aspioti is a healthcare and life sciences expert at PA Consulting. She has several years of professional experience in product innovation for medical devices and a diverse academic background in life sciences. She has worked extensively with early-stage R&D teams as a biology specialist on technology landscaping, technology evaluation, and scientific diligence. She is the co-inventor of several patents in the field of advanced wound therapies. In addition, she has helped establish and managed preclinical research programs for concept evaluation in various areas, including wound care and regenerative medicine while working with clinical groups and commercial teams to support clinical evidence and business case generation. Aspioti holds a BSc (Hons) in molecular & cellular biology from the University of Glasgow and a MSc in regenerative medicine from the University of Bath.

Paolo Siciliano is an associate partner and life sciences expert at PA Consulting, and he leads PAs work in C> globally. He has several years of experience in supporting major pharma, biotech, and medtech companies to identify, develop, and leverage new technologies to solve business needs, as well as improve their innovation and product development processes. His main areas of expertise range from technology and commercial strategy to technology development, across a number of therapeutic areas. He obtained a Ph.D. in molecular biology and worked as a senior research scientist in biotech companies in the U.K.

See the rest here:
CAR-T Beyond CGTs In Development In 2022 - BioProcess Online

Posted in Stem Cells | Comments Off on CAR-T Beyond CGTs In Development In 2022 – BioProcess Online

ADOCIA Announces First Cell Therapy Preclinical Proof of Concept of AdoShell Islets for the Treatment of Type 1 Diabetes – Business Wire

Posted: September 8, 2022 at 1:56 am

LYON, France--(BUSINESS WIRE)--Regulatory News:

Adocia (Euronext Paris: FR0011184241 ADOC), a clinical-stage biopharmaceutical company focused on the research and development of innovative therapeutic solutions for the treatment of diabetes and other metabolic diseases, announces the establishment of a first proof of concept for its AdoShell Islets implant by achieving glycemic control without insulin injections in immunocompetent diabetic rats during the 132-day study.

"Adocias results are remarkable, having successfully performed the first islet transplantation without the use of immunosuppressants in immunocompetent animals. We are delighted to be actively involved in these unprecedent results, said Dr. Karim Bouzakri, Director of CEED (European Center for the Study of Diabetes).

AdoShell Islets is an immuno-protective synthetic biomaterial containing islets of Langerhans. After implantation in diabetic animals, the islets encapsulated in AdoShell secrete insulin in response to blood glucose levels. The physical barrier formed by the AdoShell biomaterial allows the implanted cells to be invisible to the host's immune system while allowing the necessary physiological exchanges to occur for the survival and function of the islets.

This study consisted of implanting islets from allogeneic rats (Wistar) - encapsulated in AdoShell into immunocompetent diabetic rats (Lewis). The insulin secreted by the transplanted islets was measured for 132 days and no slowing of secretion was observed during the duration of the study.

At the end of the study the graft was removed, which resulted in an observable drop of insulin secretion and rise in blood sugar levels, the animals rapidly returned to its diabetic state. At the same time, the animals in the control group (diabetic rats that did not receive AdoShell Islets) were unable to control their blood sugar levels.

Additional ongoing studies in diabetic rats, with the aim to optimize the AdoShell technology, confirm these initial results, producing insulin and normalizing the glycemia in 4 diabetic rats for 80 days (study still on-going). The weight gain of the studied rats - which is also an important clinical indicator of healthy test subjects - shows that the AdoShell Islets are performing as expected. In parallel the rats in the control group are not gaining weight as expected in diabetic rats.

These results will be presented at the upcoming cell therapy session of the PODD 2022 conference held in Boston in October.

"This first proof of concept in diabetic rats validates our AdoShell technology. Our purely physico-chemical approach is unique and being not biological it gives us confidence that these remarkable results can be translated from one species to another," said Olivier Soula, Deputy-CEO and Director of R&D at Adocia.

Our priority, treating life threatening cases with cells from donors

More than 40 million people worldwide suffer from type 1 diabetes1, also known as insulin-dependent diabetes: In these patients, the beta cells of the islets of Langerhans, cells that secrete insulin, are destroyed by an autoimmune mechanism. As a result, the patient survival depends on daily injections of insulin.

Despite the use of insulin, some patients have intensely unstable diabetes characterized by extreme glycemic variability, responsible for iterative and/or severe unfelt hypoglycemia, altering the quality of life and increasing morbidity and mortality. The prognosis of this so-called "brittle" diabetes is poor, with a mortality rate between 20 to 50% over 5 years, depending on the study2. Brittle diabetes affects about 3 out of 1000 people with insulin-dependent diabetes, which represents 1000 patients in France and nearly 75 000 worldwide.

Cell therapy techniques by replacing cells that have been destroyed exist and consist in injecting the patient with islets of Langerhans taken from pancreas of donors. These techniques are practiced in many countries and in 2020 the French High Authority for Health (the HAS) gave a favorable opinion on the registration of islets transplantation on the list of procedures that can be reimbursed by the public Health Insurance. However, this technique has a major pitfall because - like any allograft - islet transplantation as practiced to date requires the concomitant use of heavy immunosuppressive treatments to avoid rejection of the transplanted cells. These immunosuppressive protocols, whose undesirable effects are widely documented (hematological anomalies, infections, and neoplasia), limit the use of transplantation techniques to patients already under immunosuppressive treatment because they are also undergoing kidney transplantation.

The first application of AdoShell Islets concerns the improvement of these techniques performed with donor pancreases and is precisely aimed at these so-called "brittle" patients so that they can benefit from them.

"Our approach is first and foremost very pragmatic: to use donor cells already used in current therapeutics and to fit into existing protocols. In this way, we hope to make a first treatment available to the most severe patients as soon as possible, said Grard Soula, Chairman and CEO of Adocia.

A technology applicable to other cellular sources with the objective of treating the greatest number of people

In parallel with the development of AdoShell Islets from donor pancreases, Adocia also aims to develop its technology from stem cells, which would ultimately make this technology possible to free itself from the limit of the number of donors and thus treat a much larger number of patients.

"We are currently working on setting up collaborations with companies that develop stem cells with an ambitious vision: to offer the best curative treatment for diabetes without requiring immunosuppressants," concluded Grard Soula, Chairman and CEO of Adocia.

During the virtual conference held on September 20th to release the financial results of the first half of 2022, we will present the AdoShell Islets program in detail to investors and shareholders, along with the company's entire program portfolio.

About Adocia

Adocia is a biotechnology company specializing in the discovery and development of therapeutic solutions in the field of metabolic diseases, primarily diabetes and obesity. The company has a broad portfolio of drug candidates based on three proprietary technology platforms:

1) The BioChaperone technology for the development of new generation insulins and products combining insulins with other classes of hormones; 2) AdOral, an oral peptide delivery technology; 3) AdoShell, an immunoprotective biomaterial for cell transplantation with a first application in pancreatic cells transplantation for patients with "brittle" diabetes.

Adocia holds more than 25 patent families.

Based in Lyon, the company has approximately 115 employees. Adocia is listed on the Euronext Paris market (Euronext: ADOC; ISIN: FR0011184241).

Disclaimer

This press release contains certain forward-looking statements concerning Adocia and its business. Such forward-looking statements are based on assumptions that Adocia considers as being reasonable. However, there can be no guarantee that the estimates contained in such forward-looking statements will be achieved, as such estimates are subject to numerous risks including those which are set forth in the Risk Factors section of the universal registration document that was filed with the French Autorit des marchs financiers on April 21, 2022 (a copy of which is available at http://www.adocia.com, in particular uncertainties that are linked to research and development, future clinical data, analyses, and the evolution of the economic context, the financial markets and the markets in which Adocia operates.

The forward-looking statements contained in this press release are also subject to risks not yet known to Adocia or not considered as material by Adocia as of this day. The occurrence of all or part of such risks could cause that actual results, financial conditions, performances, or achievements of Adocia be materially different from those mentioned in the forward-looking statements.

__________________________1 International Diabetes Federation, around 10% of all people with diabetes2 HAS juillet 2020 https://www.has-sante.fr/jcms/p_3195137/en/transplantation-d-lots-pancreatiques-rapport-d-evaluation-technologique

Go here to read the rest:
ADOCIA Announces First Cell Therapy Preclinical Proof of Concept of AdoShell Islets for the Treatment of Type 1 Diabetes - Business Wire

Posted in Stem Cells | Comments Off on ADOCIA Announces First Cell Therapy Preclinical Proof of Concept of AdoShell Islets for the Treatment of Type 1 Diabetes – Business Wire

Synthetic Mouse Embryo with Brain and Beating Heart Grown from Stem Cells – Genetic Engineering & Biotechnology News

Posted: August 30, 2022 at 2:14 am

Researchers from the University of Cambridge have harnessed mouse stem cells to create model synthetic embryos that comprise a brain, a beating heart, and the foundations of all the other organs of the body.

The team, led by Magdalena Zernicka-Goetz, PhD, mimicked natural processes, in the lab,without the use of eggs or sperm, by guiding the three types of stem cells found in early mammalian development to the point where they start interacting. By inducing the expression of a particular set of genes and establishing a unique environment for their interactions, the researchers were able to get the stem cells to talk to each other.

The stem cells self-organized into structures that progressed through the successive developmental stages until they had beating hearts and the foundations of the brain, as well as the yolk sac where the embryo develops and gets nutrients in its first weeks. Unlike other synthetic embryos, the Cambridge-developed mouse embryo models reached the point where the entire brain, including the anterior portion, began to develop. This is a further point in development than has been achieved in any other stem cell-derived model.

Our mouse embryo model not only develops a brain, but also a beating heart, all the components that go on to make up the body, said Zernicka-Goetz, who is a professor in mammalian development and stem cell biology in Cambridges department of physiology, development, and neuroscience.

The scientists say their results, which culminate from more than a decade of research that has progressively led to more and more complex embryo-like structures, could help scientists understand why some embryos fail while others go on to develop into a healthy pregnancy. Additionally, the results could be used to guide the development of synthetic human organs for transplantation. Its just unbelievable that weve got this far, Zernicka-Goetz continued. This has been the dream of our community for years, and the major focus of our work for a decade and finally weve done it.

Zernicka-Goetz, together with first author Gianluca Amadei, PhD, described their work in Nature, in a paper titled, Synthetic embryos complete gastrulation to neurulation and organogenesis, in which they concluded, these complete embryoids are a powerful in vitro model for dissecting the roles of diverse lineages and genes in development Because ETiX-embryoids capture extensive aspects of development, they provide a significant opportunity to uncover mechanisms of development and disease.

For a human embryo to develop successfully there needs to be dialogue between the tissues that will become the embryo, and the tissues that will connect the embryo to the mother. In the first week after fertilization, three types of stem cells develop: one will eventually become the tissues of the body, and the other two support the embryos development. One of these extraembryonic stem cell types will become the placenta, which connects the fetus to the mother and provides oxygen and nutrients; and the second is the yolk sac, where the embryo grows and where it gets its nutrients from in early development.

In natural development, the zygote develops into the epiblast, which will form the organism; the extraembryonic visceral endoderm (VE), which contributes to the yolk sac; and the extraembryonic ectoderm (ExE), which contributes to the placenta, the authors explained. Stem cells corresponding to these three lineages offer the possibility to completely regenerate the mammalian organism from multiple components, instead of from a single totipotent zygote. In vitro embryonic stem cells can undergo many aspects of mammalian embryogenesis, the team continued, but their developmental potential is substantially extended by interactions with extraembryonic stem cells, including trophoblast stem cells (TSCs), extraembryonic endoderm stem cells (XEN), and inducible-XEN cells (iXEN).

Many pregnancies fail at the point when the three types of stem cells begin to send mechanical and chemical signals to each other, which tell the embryo how to develop properly. So many pregnancies fail around this time, before most women realize they are pregnant, said Zernicka-Goetz, who is also a professor of biology and biological engineering at Caltech. This period is the foundation for everything else that follows in pregnancy. If it goes wrong, the pregnancy will fail.

Over the past decade, Zernicka-Goetzs group ihas been studying these earliest stages of pregnancy, in order to understand why some pregnancies fail and some succeed. The stem cell embryo model is important because it gives us accessibility to the developing structure at a stage that is normally hidden from us due to the implantation of the tiny embryo into the mothers womb, said Zernicka-Goetz. This accessibility allows us to manipulate genes to understand their developmental roles in a model experimental system.

To guide the development of their synthetic embryo, the researchers put together cultured stem cells representing each of the three types of tissue in the right proportions and environment to promote their growth and communication with each other, eventually self-assembling into an embryo. They discovered that the extraembryonic cells signal to embryonic cells by chemical signals but also mechanistically, guiding the embryos development. This period of human life is so mysterious, so to be able to see how it happens in a dishto have access to these individual stem cells, to understand why so many pregnancies fail, and how we might be able to prevent that from happeningis quite special, said Zernicka-Goetz. We looked at the dialogue that has to happen between the different types of stem cell at that timeweve shown how it occurs and how it can go wrong.

A major advance in the reported study is the ability to generate the entire brain, in particular the anterior part, which has been a major goal in the development of synthetic embryos. Our embryo model displays head-folds with defined forebrain and midbrain regions the investigators noted. The teams previous studies had used the same component cells to develop into embryos at a slightly earlier stage. Now, by pushing development just one day further, they say that their model is the very first to signal development of the anterior, and in fact the whole, brain.

This opens new possibilities to study the mechanisms of neurodevelopment in an experimental model, said Zernicka-Goetz. In fact, we demonstrate the proof of this principle in the paper by knocking out a gene already known to be essential for formation of the neural tube, precursor of the nervous system, and for brain and eye development. In the absence of this gene, the synthetic embryos show exactly the known defects in brain development as in an animal carrying this mutation. This means we can begin to apply this kind of approach to the many genes with unknown function in brain development.

As the authors noted, Importantly, we were able to replicate the consequences of Pax6 knockout in neurulating embryoids, which illustrates the potential of this complete embryo model to dissect the genetic factors that regulate development without the need of experimental animals.

In conclusion, they stated, Here, we show that we can assemble mouse embryonic and extraembryonic stem cells to form an embryo model that develops the brain, neural tube, heart, foregut, somite, allantois, primordial germ cells, and yolk sac structures. This embryo model is able to achieve this entirely through self-organization of these three stem cell types, without the need to provide any additional external signalling cues.

While the current research was carried out in mouse models, the researchers are developing similar human models, potentially enabling the development specific organ types that could help scientists understand mechanisms behind processes that would be otherwise impossible to study in real embryos. At present, under UK law, human embryos can be studied in the laboratory only up to the fourteenth day of development.

If the methods developed by Zernicka-Goetzs team are shown to be successful with human stem cells, they could feasibly be used to guide development of synthetic organs as human transplants. There are so many people around the world who wait for years for organ transplants, said Zernicka-Goetz. What makes our work so exciting is that the knowledge coming out of it could be used to grow correct synthetic human organs to save lives that are currently lost. It should also be possible to affect and heal adult organs by using the knowledge we have on how they are made.

This is an incredible step forward and took 10 years of hard work of many of my team membersI never thought wed get to this place. You never think your dreams will come true, but they have.

The newly reported work comes weeks after the publication, in Cell, of a study by a team led by co-author Jacob Hanna, PhD, at the Weizmann Institute. James Briscoe, PhD, principal group leader, assistant research director, Francis Crick Institute, said, that similar to the research recently reported by Hanna and colleagues, the study by Zernicka-Goetz and colleagues represented valuable proof of concept demonstration that a synthetic mouse embryo-like structure can be assembled from stem cells. By combining these cells together, the study shows that it is possible to coax the development of something that resembles a mouse embryo at a stage when the main organs of the body are beginning to be established, including the nervous system, heart, and gut, Briscoe said.

However, Briscoe pointed out that that formation of the synthetic embryos was very inefficient, that even the successful synthetic embryos appeared not as well organized as natural embryos, and that they didnt develop beyond what would be day 8.5 of normal embryonic development, which is just under halfway through a normal mouse pregnancy.

This emphasizes how much we still have to learn about how embryos build themselves, he noted. The technique reported in this study is a promising approach to provide new insights into how mammalian embryos organize and construct organs. Nevertheless, the study has broad implications as, although the prospect of synthetic human embryos still requires further research (as human embryos are not identical to mouse embryos), now is a good time to engage in wider discussions about the legal and ethical implications of such research.

See the original post here:
Synthetic Mouse Embryo with Brain and Beating Heart Grown from Stem Cells - Genetic Engineering & Biotechnology News

Posted in Stem Cells | Comments Off on Synthetic Mouse Embryo with Brain and Beating Heart Grown from Stem Cells – Genetic Engineering & Biotechnology News

Utilization of Modified Induced Pluripotent Stem Cells as the Advance | OPTH – Dove Medical Press

Posted: August 30, 2022 at 2:14 am

Introduction

Glaucoma is one of the optic neuropathy disorders characterized by the progressive degeneration of retinal ganglion cells (RGC), which eventually lead to cupping of the optic disc and decreased visual field.1 Glaucoma is also closely related to an increase in intraocular pressure caused by the damage of trabecular meshwork (TBM), which results in optic nerve damage, characterized by the loss of retinal ganglion cells.2,3 Globally, in 2020, more than 76 million people are suffering from glaucoma, and it is expected to increase to 111.8 million people by 2040.4,5 Glaucoma is also a severe and complex medical problem because it often causes blindness. According to the World Health Organization (WHO), the most common causes of blindness are cataracts (51%), followed by glaucoma (8%), and age-related macular degeneration (5%).6 This data shows that glaucoma is the worlds second most common cause of blindness after cataracts. Symptoms that are often asymptomatic at an early stage and the low public awareness have contributed to the disorders seriousness.

Handling and treating glaucoma cases is difficult, especially because no therapy can cure glaucoma. Current treatment, both medical and surgical, is focused solely on lowering intraocular pressure. Treatment of glaucoma cases should also be carried out for life to maintain normal intraocular pressure and prevent the progression of intraocular damage due to glaucoma.7 Based on these problems, innovation is needed to handle glaucoma effectively. Besides, solutions are also required to repair the damage to retinal ganglion cells in glaucoma. One of the therapies that researchers are trying to take advantage of is stem cell therapy, a technology where cells can develop into many specific cells desired.8 In cases of glaucoma or optic neuropathies, damaged RGCs can be replaced with new ones grown from stem cells.9 Another option for RGC regeneration is to use retinal stem cells to regenerate RGCs. Indeed, stem cell therapy relies on exogenous stem cell sources due to their limited availability. Currently, many stem cell therapies for eye diseases that are created and studied are limited to treating the damage of photoreceptors and retinal pigment epithelium. iPSC-derived RGCs can serve as an excellent model for formulating approaches to promote de novo-generated RGCs to connect with their targets. Therefore, researchers have been looking into the potential use of modified stem cell therapy to treat the intraocular injury in glaucoma cases.10

This review aims to synthesize and prove the efficacy and further modification of this method so that it can be eligible for treatment and can also give data collection for the scientific community. This systematic review is expected to provide detailed information regarding the possible applications of modified stem cell therapy in treating intraocular damage in glaucoma patients.

In the present literature review, literature regarding the potential utilization of stem cells as an advanced therapy for intraocular glaucomatous damage was searched. The stages of this literature review include five steps: i) identifying the research question, ii) identifying relevant studies, iii) study selection, iv) charting the data, and v) summarizing and reporting the results.

This literature review was conducted to answer the following research questions:

The literature search was carried out from January to February 2021. Keywords and synonyms used to conduct literature searches related to the research question are attached in Table 1. Boolean operators (OR, AND, NOT) combine keywords when searching for literature. The search was conducted on seven online databases, namely PubMed, ScienceDirect, ProQuest, EBSCOhost, SAGE, Clinicalkey, and Scopus.

Table 1 Keywords That Were Used in the Database Search

The inclusion criteria for the literature search consisted of journals published in English and journals published in the last ten years. The exclusion criteria for selected studies consisted of journals that were not fully accessible due to the limited facilities owned as supporting access. We thoroughly screened the titles and abstracts of the studies obtained to suit the purpose of this literature review. Abstracts that were not relevant to the research objectives were excluded. Then a full article screening was carried out from the selected abstracts to identify whether the full article was suitable for the research objectives and whether the full article could be used to answer research questions.

Information obtained from all selected study articles is then displayed in the charting table The information displayed includes the author, year of publication, study objectives, location, study design, inclusion and exclusion criteria, results, and conclusions.

The researcher did not assess the quality of the selected articles because this study was only a literature review. The data from selected studies are reported to produce recommendations for further research regarding the use of stem cell therapy in glaucoma cases.

Based on the literature search that has been conducted, a total of 2262 studies and abstracts were included in the journal screening process at an early stage. From this screening process, 362 duplicate articles were excluded from the selection. The remaining 1900 articles then entered the abstract eligibility screening stage. Only 53 articles were selected, while 1879 other articles were excluded. Of the 53 articles, 18 articles appeared relevant to the study and met the inclusion criteria for review throughout the study. Meanwhile, 35 other studies were excluded because the focus in these studies did not match the objectives of this literature review. After assessing the full articles, six studies met the inclusion criteria in this literature review (Figure 1).

Figure 1 Flow diagram of the literature review process.

In Table 2, a summary of the characteristics of the selected studies is presented. The data used from selected studies include research objectivity, study design, results, outputs, and conclusions from the study. Of all the selected studies, there were six studies that had experimental methods. Almost all studies have the aim of evaluating and proving the potential of using stem cells to replace damaged tissue and restore and restore the function of damaged eye tissue, particularly due to degenerative processes such as disease of the retina or glaucoma.

Table 2 Results Summary of the Characteristics of the Selected Studies

Glaucoma is characterized by the degeneration of retinal ganglion cells. Based on the pathophysiology, glaucoma can be divided into two categories, namely open-angle glaucoma and closed-angle glaucoma. In patients with open-angle glaucoma, there is increased resistance to the aqueous humors outflow through the trabecular meshwork. This increased resistance is often caused by apoptosis and senescence of trabecular meshwork cells with increasing age.15 Degradation and abnormalities of the cytoskeleton arrangement of trabecular meshwork cells resulting in thickening of the drainage pathways and abnormal extracellular matrix deposition also worsen trabecular meshwork function in open-angle glaucoma.16 In closed-angle glaucoma, the aqueous humor cannot reach the trabecular meshwork due to obstruction.17 Examples of obstructions that often cause closed-angle glaucoma are anterior synechiae, the attachment of the iris to the trabecular meshwork, and posterior synechiae, where the iris is attached to the lens. This adhesion causes the aqueous humor to fail to reach the drainage system and the trabecular meshwork.18

Glaucoma is closely related to increased intraocular pressure, which is determined by the balance between the production of aqueous humor by the ciliary body and the drainage of the aqueous humor through the trabecular meshwork. The disturbance of the balance between production and drainage increases the humor Aquos, which at a later stage can increase the intraocular pressure.19 Studies have shown a link between increased intraocular pressure and retinal ganglion cell death. This study has also proven that the longer the intraocular pressure increases, the higher the degree of retinal ganglion cell damage.20 However, data show as many as 3040% of patients with glaucoma have normal intraocular pressure. One of the causes of glaucoma at normal intraocular pressure is a decrease in neurotrophic factors needed in the maintenance of neurons in the optic nerve. Neurotrophic factors are required to maintain retinal ganglion cells, including brain-derived neurotrophic factor (BDNF), ciliary neurotrophic factor (CNTF), and cell line-derived neurotrophic factor.21 Furthermore, microcirculation disorders, changes in immune system conditions, and increased levels of oxidative stress can also cause glaucoma at normal intraocular pressure.21

Stem cells are cells with the ability to differentiate and form all tissues in the human body. They are one of the potential therapies used in cases that require tissue repair and regeneration, one of which is glaucoma. For a cell to be called a stem cell, it must have two essential characteristics. The first one is the stem cell must produce offspring with the exact features the cell originates from, and the second one, the stem cell must be able to differentiate into the specific cell desired.22 There are two types of stem cells found in multicellular organisms, including humans. The first stem cells are embryonic stem cells or multipotent cells found in blastocysts, while the second stem cells are adult stem cells or pluripotent cells that can be found in a wide variety of adult tissues.23

Research has also succeeded in inducing adult cells to return to the pluripotent stage using molecular manipulation. The cells produced by this molecular manipulation are then called induced pluripotent stem cells (iPS).24 Most iPS manufacturing uses viruses such as retroviruses and lentiviruses to carry genes encoding transcription factors to adult cells to be modified. This gene will then undergo transcription and translation into a protein capable of inducing the adult cell nucleus to return to an embryonic state.25

An important concept that needs attention in stem cell therapy is how to induce stem cells to become the desired differentiated cells.26 It is necessary so that the cells can be used to treat various diseases, including glaucoma. We can further achieve differentiation of stem cells into specific desired cells by adding various growth factors and signaling pathways to resemble the conditions of their original development.27

The research conducted successfully isolates cultures and confirms that the trabecular meshwork stem cells around the Schwalbe line are multipotent with the ability to differentiate into a wide variety of cells, including trabecular meshwork cells adipocytes osteocytes, and chondrocytes.28 Other studies have also been able to induce stem cells on the Schwalbe line trabecular meshwork to proliferate and differentiate into photoreceptors under certain conditions.29 Apart from trabecular meshwork stem cells, other stem cells that can differentiate into functional meshwork trabecular cells are adipose-derived stem cells (ADSC), mesenchymal stem cells (MSC), and iPS. iPS cells can also differentiate into trabecular meshwork cells after culturing the extracellular matrix with cell-derived trabecular meshwork. The success of a wide variety of stem cells to differentiate into functional meshwork trabecular cells provides a more effective alternative to cutting-edge therapy in treating glaucoma, especially open-angle glaucoma.3

One of the stem cell therapies successfully applied and able to regenerate damaged retinal ganglion cells is iPS cell therapy. This therapy uses induced adult fibroblasts to return to pluripotent cells using four transcription factors, namely Oct3/4, Sox2, Klf4, and c-Myc. The results of the iPS are pluripotent cell colonies that are morphologically similar to ESCs, which are able to differentiate into the three germ cell layers.30

Because iPS can be programmed from the patients somatic cells, this therapy can maintain the unique genome of each individual. Currently, various modifications to the iPS therapy have been made to increase its acceptability and effectiveness of iPS therapy. One of them is the use of plasmid vectors and miRNA instead of retroviruses to avoid mutagenesis of the adult cells used.31,32

One of the significant challenges in stem cell therapy is to achieve the differentiation of stem cells into the desired cells, in this case, the differentiation of stem cells to retinal ganglion cells. Usually, in vivo, the differentiation of stem cells into retinal ganglion cells is regulated by several transcription factors such as Ath5, Brn3, and Notch. The transcription factors Ath5 and Brn3 play a vital role in the differentiation of retinal ganglion cells, and their levels are increased in the process of eye development.33 Meanwhile, Notch is a negative regulator of retinal ganglion cell differentiation, and its levels are decreased in normal eye development. Therefore, the addition of the transcription factors Ath5 and Brn3 and the Notch antagonist is a strategy to differentiate retinal ganglion cells from stem cells.34 Apart from transcription factors, various neurotrophic pathways and factors have been identified in the differentiation of stem cells into retinal ganglion cells. These pathways consist of fibroblast growth factor (FGF), insulin-like growth factor (IGF), bone morphogenetic protein (BMP), nodal, and Wnt signaling pathways. All of these pathways regulate retinal development, whereas FGF and IGF provide positive regulation. Meanwhile, BMP, nodal, and Wnt signaling pathways provide negative regulation.35

Another major challenge in the clinical application of stem cell therapy in glaucoma sufferers is that not only do the stem cells successfully differentiate into retinal ganglion cells, but they must also be able to reach the central nervous system.36 Modifications must be made so that new retinal ganglion cells can reach the visual cortex of the cerebrum. Recent research has found that a combination of genetic modification and stimulation of the signaling pathway stimulates regeneration of the optic nerve until it reaches the central nervous system. The addition of ephrin molecules, proteoglycans, cell-adhesion molecules, and semaphorin is able to guide the axons of the developing retinal ganglion cells to reach the optic chiasm.13 Meanwhile, the addition of cadherin, ephrin, and the Wnt signaling pathway can guide and stimulate synapse formation in the superior colliculus and the visual cortex.12,37

In addition, because of the adverse intraocular environment in glaucoma, stem cell therapy needs to be combined with neuroprotective compounds. It is also associated with a decrease in neurotrophic factors required to maintain neurons and causes progression of retinal ganglion cell damage in glaucoma sufferers. Therefore, the addition of BDNF and other neurotrophic factors such as glial cell-derived neurotrophic factor (GDNF) and ciliary neurotrophic factor (CNTF) should be considered for combined stem cell therapy.38

The stem cells are used in cases of glaucoma, which require repair and regeneration of trabecular meshwork cells and retinal ganglion cells. iPS has been shown the ability to differentiate to replace damaged trabecular meshwork cells and retinal ganglion cells in glaucoma. Some modifications are required so that stem cells that have differentiated into trabecular meshwork cells and retinal ganglion cells can reach the central nervous system. These modifications include the addition of ephrin molecules, proteoglycans, cell-adhesion molecules, semaphorin, cadherin, and the Wnt signaling pathway. The combination of stem cells with neuroprotective factors such as BDNF, GDNF, and CNTF also needs to be considered to maintain neuronal maintenance and inhibit the progression of cell damage.

The development of new stem cell technologies not only paves the way for us to gain a better understanding of the biology associated with glaucoma and create models for the development of new drugs, but it also opens the door to the prospect of cell-based therapies that can help patients regain their vision. More specifically in relation to the field of glaucoma, there have been recent developments in the process of developing protocols for the differentiation of stem cells into trabecular meshwork and retinal ganglion cells. Further research on the effectiveness of using modified stem cells as a therapy for glaucoma and in vivo research can be carried out immediately so that clinical trials can be carried out, which in turn can be used by the community to control symptoms and reduce blindness due to glaucoma.

The authors report no conflicts of interest in this work.

1. Choudhari NS, Neog A, Fudnawala V, George R. Cupped disc with normal intraocular pressure: the long road to avoid misdiagnosis. Indian J Ophthalmol. 2011;59(6):491. doi:10.4103/0301-4738.86320

2. Braunger BM, Ademoglu B, Koschade SE, et al. Identification of adult stem cells in Schwalbes line region of the primate eye. Invest Ophthalmol Vis Sci. 2014;55(11):7499. doi:10.1167/iovs.14-14872

3. Abu-Hassan DW, Li X, Ryan EI, Acott TS, Kelley MJ. Induced pluripotent stem cells restore function in a human cell loss model of open-angle glaucoma. Stem Cells. 2015;33(3):751761. doi:10.1002/stem.1885

4. Cook C, Foster P. Epidemiology of glaucoma: whats new? Can J Ophthalmol. 2012;47(3):223226. doi:10.1016/j.jcjo.2012.02.003

5. Hapsari DM. The Relationship Between Family Support and Adherence in Care in Glaucoma Clients in the Working Area of the Balung Health Center. Jember Regency. Uni; 2017.

6. World Health Organization. Global data on visual impairments 2010; 2012. https://www.iapb.org/wp-content/uploads/GLOBALDATAFINALforweb.pdf. Accessed August 24, 2022.

7. Harasymowycz P, Birt C, Gooi P, et al. Medical management of glaucoma in the 21st century from a canadian perspective. J Ophthalmol. 2016;2016:122. doi:10.1155/2016/6509809

8. Romito A, Cobellis G. Pluripotent stem cells: current understanding and future directions. Stem Cells Int. 2016;2016:120. doi:10.1155/2016/9451492

9. Cen L-P, Ng TK. Stem cell therapy for retinal ganglion cell degeneration. Neural Regen Res. 2018;13(8):1352. doi:10.4103/1673-5374.235237

10. ner A. Stem cell treatment in retinal diseases: recent developments. Turk J Ophthalmol. 2018;48(1). doi:10.4274/tjo.89972

11. Manuguerra-Gagn R, Boulos PR, Ammar A, et al. Transplantation of mesenchymal stem cells promotes tissue regeneration in a glaucoma model through laser-induced paracrine factor secretion and progenitor cell recruitment. Stem Cells. 2013;31(6):11361148. doi:10.1002/stem.1364

12. Osterhout JA, Josten N, Yamada J, et al. Cadherin-6 mediates axon-target matching in a non-image-forming visual circuit. Neuron. 2011;71(4):632639. doi:10.1016/j.neuron.2011.07.006

13. De lima S, Koriyama Y, Kurimoto T, et al. Full-length axon regeneration in the adult mouse optic nerve and partial recovery of simple visual behaviors. Proc Natl Acad Sci U S A. 2012;109(23):91499154. doi:10.1073/pnas.1119449109

14. Deng F, Chen M, Liu Y, et al. Stage-specific differentiation of iPSCs toward retinal ganglion cell lineage. Mol Vis. 2016;22:536.

15. Pulliero A, Seydel A, Camoirano A, Sacc SC, Sandri M, Izzotti A. Oxidative damage and autophagy in the human trabecular meshwork as related with ageing. PLoS One. 2014;9(6):e98106. doi:10.1371/journal.pone.0098106

16. Porter KM, Epstein DL, Liton PB. Up-regulated expression of extracellular matrix remodeling genes in phagocytically challenged trabecular meshwork cells. PLoS One. 2012;7(4):e34792. doi:10.1371/journal.pone.0034792

17. Lai J, Choy BNK, Shum JWH. Management of primary angle-closure glaucoma. Asia Pac J Ophthalmol. 2016;5(1):5962. doi:10.1097/APO.0000000000000180

18. Sun X, Dai Y, Chen Y, et al. Primary angle closure glaucoma: what we know and what we dont know. Prog Retin Eye Res. 2017;57:2645. doi:10.1016/j.preteyeres.2016.12.003

19. Gemenetzi M, Yang Y, Lotery AJ. Current concepts on primary open-angle glaucoma genetics: a contribution to disease pathophysiology and future treatment. Eye. 2012;26(3):355369. doi:10.1038/eye.2011.309

20. Khan Kareem A, Tse DY, van der Heijden ME, et al. Prolonged elevation of intraocular pressure results in retinal ganglion cell loss and abnormal retinal function in mice. Exp Eye Res. 2015;130. doi:10.1016/j.exer.2014.11.007

21. Almasieh M, Wilson AM, Morquette B, Cueva Vargas JL, Di Polo A. The molecular basis of retinal ganglion cell death in glaucoma. Prog Retin Eye Res. 2012;31(2):152181. doi:10.1016/j.preteyeres.2011.11.002

22. Cao J, Ng ES, Mcnaughton D, et al. The characterisation of pluripotent and multipotent stem cells using Fourier transform infrared microspectroscopy. Int J Mol Sci. 2013;14(9):1745317476. doi:10.3390/ijms140917453

23. Makhani K, Ali SM, Yousuf S, Siddiqui S. Therapeutic potential of totipotent, pluripotent and multipotent stem cells. MOJ Cell Sci Rep. 2015;2(5):00041. doi:10.15406/mojcsr.2015.02.00041

24. Zaveri L, Dhawan J. Cycling to meet fate: connecting pluripotency to the cell cycle. Front Cell Dev Biol. 2018;6. doi:10.3389/fcell.2018.00057

25. Wattanapanitch M. Recent updates on induced pluripotent stem cells in hematological disorders. Stem Cells Int. 2019;2019:115. doi:10.1155/2019/5171032

26. Kolios G, Moodley Y. Introduction to stem cells and regenerative medicine. Respiration. 2012;85(1):310. doi:10.1159/000345615

27. Biehl JK, Russell B. Introduction to stem cell therapy. J Cardiovasc Nurs. 2009;24(2):98103; quiz 1045. doi:10.1097/JCN.0b013e318197a6a5

28. Tay CY, Sathiyanathan P, Chu SWL, Stanton LW, Wong TT. Identification and characterization of mesenchymal stem cells derived from the trabecular meshwork of the human eye. Stem Cells Dev. 2012;21(9):13811390. doi:10.1089/scd.2011.0655

29. Nadri S, Yazdani S, Arefian E, et al. Mesenchymal stem cells from trabecular meshwork become photoreceptor-like cells on amniotic membrane. Neurosci Lett. 2013;541:4348. doi:10.1016/j.neulet.2012.12.055

30. Omole AE, Fakoya AOJ. Ten years of progress and promise of induced pluripotent stem cells: historical origins, characteristics, mechanisms, limitations, and potential applications. PeerJ. 2018;2018. doi:10.7717/peerj.4370

31. Miyoshi N, Ishii H, Nagano H, et al. Reprogramming of mouse and human cells to pluripotency using mature microRNAs. Cell Stem Cell. 2011;8(6):633638. doi:10.1016/j.stem.2011.05.001

32. Ikushima S, Ono R, Fukuda K, Sakayori M, Awano N, Kondo K. Trousseaus syndrome: cancer-associated thrombosis. Jpn J Clin Oncol. 2016;46(3):204208. doi:10.1093/jjco/hyv165

33. Ji SL, Tang SB. Differentiation of retinal ganglion cells from induced pluripotent stem cells: a review. Int J Ophthalmol. 2019;12(1):152160. doi:10.18240/ijo.2019.01.22

34. Li L, Chen LP, Liu QH. Effect of the notch signaling pathway on retinal ganglion cells and its neuroprotection in rats with acute ocular hypertension. Int J Ophthalmol. 2018;11(2). doi:10.18240/ijo.2018.02.05

35. Ritchey ER, Zelinka CP, Tang J, Liu J, Fischer AJ. The combination of IGF1 and FGF2 and the induction of excessive ocular growth and extreme myopia. Exp Eye Res. 2012;99(1):116. doi:10.1016/j.exer.2012.03.019

36. Georgiev D. Photons do collapse in the retina not in the brain cortex: evidence from visual illusions. NeuroQuantology. 2011;9(2). doi:10.14704/nq.2011.9.2.403

37. He CW, Liao CP, Pan CL. Wnt signalling in the development of axon, dendrites and synapses. Open Biol. 2018;8(10):180116. doi:10.1098/rsob.180116

38. Mysona BA, Zhao J, Bollinger KE. Role of BDNF/TrkB pathway in the visual system: therapeutic implications for glaucoma. Expert Rev Ophthalmol. 2017;12(1):6981. doi:10.1080/17469899.2017.1259566

Visit link:
Utilization of Modified Induced Pluripotent Stem Cells as the Advance | OPTH - Dove Medical Press

Posted in Stem Cells | Comments Off on Utilization of Modified Induced Pluripotent Stem Cells as the Advance | OPTH – Dove Medical Press

Rise In Number Of CROS In Various Regions Such As Europe Is Expected To Fuel The Growth Of Induced Pluripotent Stem Cell Market At An Impressive CAGR…

Posted: August 30, 2022 at 2:14 am

Rise In Research And Development Projects In Various Regions Such As East Asia, South Asia Are Expected To Offer An Opportunity Of US $ 0.5 Bn In 2022-2026 Period.

Fact.MR A Market Research and Competitive Intelligence Provider: The global induced pluripotent stem cell (iPSC) market was valued at US $ 1.8 Bn in 2022, and is expected to witness a value of US $ 2.3 Bn by the end of 2026.

Moreover, historically, demand for induced pluripotent stem cells had witnessed a CAGR of 6.6%.

Rise in spending on research and development activities in various sectors such as healthcare industry is expected to drive the adoption of human Ips cell lines in various applications such as personalized medicine and precision.

Moreover, increasing scope of application of human iPSC cell lines in precision medicine and emphasis on therapeutic applications of stem cells are expected to be driving factors of iPSC market during the forecast period.

Surge in government spending and high awareness about stem cell research across various organizations are predicted to impact demand for induced pluripotent stem cells. Rising prevalence of chronic diseases and high adoption of stem cells in their treatment is expected to boost the market growth potential.

For more insights into the Market, Request Brochure of this Report

https://www.factmr.com/connectus/sample?flag=B&rep_id=7298

Besides this, various cells such as neural stem cells, embryonic stem cells umbilical cord stem cells, etc. are anticipated to witness high demand in the U.S. due to surge in popularity of stem cell therapies.

Key Takeaways:

Growth Drivers:

For Comprehensive Insights Ask An Analyst Here

https://www.factmr.com/connectus/sample?flag=AE&rep_id=7298

Key Restraints:

Competitive Landscape:

Many key players in the market are increasing their investments in R&D to provide offerings in stem cell therapies, which are gaining traction for the treatment of various chronic diseases.

For instance:

More Valuable Insights on Induced Pluripotent Stem Cell Market

Get Customization on this Report for Specific Research Solutions

https://www.factmr.com/connectus/sample?flag=RC&rep_id=7298

Explore Fact.MRs Coverage on the Healthcare Domain

Human Umbilical Vein Endothelial Cells (HUVEC) Market - According to Fact.MR, the oncology segment is anticipated to accrue highly lucrative gains to the human umbilical vein endothelial cells (HUVEC) market, attributed to their purported effectiveness in targeting malignant growth. Simultaneously, uptake across tissue culture is also likely to widen in the future.

Cardiac Mapping System Market - The advent of cardiac mapping systems enabled electrophysiologists to target and treat complex arrhythmias more effectively than ever. According to the Centers for Disease Control and Prevention, more than 600,000 people die of heart disease in the United States, every year, making it essential for the medical professionals to take strong awareness initiatives to tell people about the availability of cardiac mapping technology, in a move to control the ever-growing cardiac deaths.

Cardiac Ablation Technologies Market - A surge in atrial fibrillation cases is proving to be a growth generator as cardiac ablation technology is prominently used for its treatment. Revenue across the radiofrequency segment is expected to gather considerable momentum, anticipated to account for over50%of global revenue.

Cardiac Patch Monitor Market - A cardiovascular monitor screen is a gadget that you control to record the electrical movement of your heart (ECG). This gadget is about the size of a pager. Cardiac patch monitors are utilized when you need long haul observing of side effects that happen not exactly day by day.

Cardiac Resynchronization Therapy Market - Newly-released Cardiac Resynchronization Therapy industry analysis report by Fact.MR shows that global sales of Cardiac Resynchronization Therapy in 2021 were held atUS$ 5.7 Bn. With7.9%, the projected market growth during 2022-2032 is expected to be slightly lower than the historical growth. CRT-Defibrillator is expected to be the higher revenue-generating product, accounting for an absolute dollar opportunity of nearlyUS$ 4 Bnduring 2022 2032.

About Fact.MR

Fact.MR is a market research and consulting agency with deep expertise in emerging market intelligence. Spanning a wide range from automotive & industry 4.0 to healthcare, technology, chemical and materials, to even the most niche categories. 80% of Fortune 1000's trusts us in critical decision making. We provide both qualitative and quantitative research, spanning market forecast, market segmentation, competitor analysis, and consumer sentiment analysis.

Contact:

Mahendra SinghUS Sales Office11140Rockville PikeSuite 400Rockville, MD20852United StatesTel: +1 (628) 251-1583E:sales@factmr.com

Follow Us:LinkedIn | Twitter

Read this article:
Rise In Number Of CROS In Various Regions Such As Europe Is Expected To Fuel The Growth Of Induced Pluripotent Stem Cell Market At An Impressive CAGR...

Posted in Stem Cells | Comments Off on Rise In Number Of CROS In Various Regions Such As Europe Is Expected To Fuel The Growth Of Induced Pluripotent Stem Cell Market At An Impressive CAGR…

Brush Up: Hematopoietic Stem Cells and Their Role in Development and Disease Therapy – The Scientist

Posted: August 30, 2022 at 2:14 am

What Are Hematopoietic Stem Cells and Why Are They Important? Hematopietic stem cells (HSCs) are multipotent cells found in the blood and bone marrow with the ability to self-renew and differentiate into multiple cell types during bone marrow hematopoiesis. Clinicians use HSCs to replace or repopulate a patients blood as a form of regenerative medicine. Research into HSC development and aging facilitates better in vitro HSC expansion and broadens their potential for disease treatment, enhancing their clinical therapeutic effects.

How Hematopoietic Stem Cells DevelopHSCs begin their development during embryogenesis in the dorsal aortic tissue and are additionally found in the placenta, yolk sac, and fetal liver. This fetal hematopoiesis process is necessary to produce the blood cells required for tissue development while generating a pool of undifferentiated HSCs. At birth, these HSCs migrate into and populate the newly-formed bone marrow and maintain a steady state of self-renewal and differentiation.1 HSCs function by producing red blood cells, platelets, and white blood cells throughout life, maintaining their levels following bleeding and infection. HSCs generally give rise to partly differentiated but proliferative progenitors, which differentiate into mature cells. Because of this process, true HSCs are relatively rare in the human body.2

Using Hematopoietic Stem Cells for Research and TreatmentHematopoietic stem cell transplantsFor more than 60 years, hematopoietic stem cell transplants (HSCTs) have been the most common form of HSC therapy, and are a standard option for treating hematologic malignancies, immunodeficiency, and defective hematopoiesis disorders. HSCs are now derived from multiple sources, such as peripheral and cord blood and bone marrow. Before transplantation, the receiving patient must undergo severe immunosuppressive procedures to prevent rejection of the new stem cells.3

Hematopoietic stem cell isolationThe most common HSC isolation method involves removing blood cells from plasma using density gradient centrifugation followed by magnetic bead isolation using the CD34+ surface marker, a general marker for all hematopoietic progenitors. Using flow cytometry, scientists sort specific HSC cell types based on common cell surface markers.4 Clinicians then intravenously infuse these cells into the receiver patients marrow where they engraft and repopulate the blood and immune system. In blood cancers such as leukemias and lymphomas, restoration of the blood system by HSCT allows patients to receive high-dose chemotherapy treatments, ridding them of malignant cells. In patients with red blood cell conditions where continuous blood transfusions are not an option, such as thalassemia major, HSCT results in 80 percent disease-free survival.5

Hematopoietic stem cells in gene and tissue regeneration therapyBone marrow hematopoietic stem cells also differentiate into cells of other lineages, such as endothelial cells, cardiomyocytes, neural cells, and hepatocytes, in a process called transdifferentiation. Because adult stem cells are rare, understanding the mechanisms behind HSC transdifferentiation could provide an additional source of tissue-specific multipotent cells and influence future clinical methods for tissue regeneration. HSCs can also help repair injured organs by releasing regenerative cytokines and recruiting cells to the damage site.5 Some of the latest advances in HSC therapeutic research involve using methods such as CRISPR for correcting genetically-defective HSCs. These methods will allow a patient to receive their own genetically-compatible (syngeneic) HSCs. These are called allogeneic transplants and are more effective at avoiding graft-versus-host disease, a condition where transplants from a donor are rejected by the recipients body, leading to an immune response against other tissues and organs. Creating genetically-corrected induced pluripotent stem cells (iPSCs) from patient skin tissues and differentiating them into HSCs has also been an active area of research, although current methods remain costly and time-consuming.6 Further research is necessary to take advantage of these remarkable multipotent cells in disease therapies.

References

1. H.K. Mikkola, S.H. Orkin, The journey of developing hematopoietic stem cells, Development, 133(19):3733-44, 2006.

2. G.M. Crane et al., Adult haematopoietic stem cell niches, Nat Rev Immunol, 17(9):573-90, 2017.

3. S. Giralt, M.R. Bishop, Principles and overview of allogeneic hematopoietic stem cell transplantation, Cancer Treat Res, 144:1-21, 2009.

4. B. Kumar, S.S. Madabushi, Identification and isolation of mice and human hematopoietic stem cells, Methods Mol Biol, 1842:55-68, 2018.

5. J.Y. Lee, S.H. Hong, Hematopoietic stem cells and their roles in tissue regeneration, Int J Stem Cells, 13(1):1-12, 2020.

6. S. Demirci et al., Hematopoietic stem cells from pluripotent stem cells: Clinical potential, challenges, and future perspectives, Stem Cells Transl Med, 9(12):1549-57, 2020.

Go here to see the original:
Brush Up: Hematopoietic Stem Cells and Their Role in Development and Disease Therapy - The Scientist

Posted in Stem Cells | Comments Off on Brush Up: Hematopoietic Stem Cells and Their Role in Development and Disease Therapy – The Scientist

Regenerative Properties of the Newborn Heart Offers Hope for Those With Congenital Heart Disease – The Epoch Times

Posted: August 30, 2022 at 2:14 am

Researchers from the Murdochs Children Research Institute (MCRI) are developing new treatments for congenital heart disease that could enable children born with birth defects can regenerate the damaged organ.

In 2011, Prof. Enzo Porello, who is nowhead of the Heart Regeneration Laboratory at the MCRI,demonstrated the regenerative properties of newborn mouse hearts at the University of Texas Southwestern Medical Centre. Prior to this research, the capacity of mammalian hearts to regenerate was a debated topic.

This sort of changed our thinking of what was possible in terms of stimulating the human heart to regenerate itself following damage, such as a heart attack, Porrello said, reported theAustralian. And I guess this also fuelled my own interest in my subsequent career in the area of regenerative medicine.

After hearing about cases where newborns recovered from massive heart attacks, Porello began to explore the regenerative properties of human newborn hearts.

In 2017, Porello and Prof. James Hudson manufactured living and beating heart tissues from stem cells in a laboratory at the University of Queensland.

Porello said that although other scientists had grown heart muscle cells from stem cells, nobody had grown the cells as miniature complex three-dimensional tissues. Additionally, they were not able to grow such tissues in a format compliant to drug development, he said.

And thats really the technological breakthrough that we were able to make.

According to the Australian Institute of Health and Welfare, approximately 9 out of every 1,000 babies born around the world will be born with congenital heart disease. In Australia, it is estimated that 2,400 babies are born with congenital heart disease annually, while in America, nearly one percent of all babies born are estimatedby the Centre For Disease Control to have the condition.

Porello said that, at the moment, if a child develops heart failure and doesnt respond to standard frontline therapies, a heart transplant is their only option. Children in this situation are put on a transplant waiting list, and whilst waiting for a heart to become available, they are put on mechanical support.

Heart transplantation is limited by organ donor availability, and its also limited by the need for lifelong immunosuppression in those patients, Porello said.

And so if were able to develop these bioengineered heart tissues from stem cells, this could potentially prevent or delay the need for heart transplantation in these very unwell individuals with end-stage heart failure.

Porello said that the ultimate goal of his research is to harness the self-repairing capacity of the newborn heart and to develop drugs that waken the hearts dormant regenerative abilities so that the organ may repair itself after damage.

I would say that based on recent studies in the field in the past 10 years since we first made our discovery in mice, we are certainly getting closer, he said.

There is sort of proof of concept that this is possible now, at least in mice, and the question is whether or not we can now make that a therapeutic reality in humans.

The first step in creating these complex heart tissues is attaching special molecules to stem cells; these molecules trigger the cells to morph into heart muscle tissue. The heart tissues are then developed in a plastic culture dish that consists of 96 tiny wells.

The geometry of the well is designed in such a way that the heart tissues spontaneously form when the heart muscle cells are inserted into the well, Porrello said.

He said that within each well of the device are tiny elastic micropillars; the pillars function as elastic cantilevers since they are attached to the dish at only one end and extend horizontally to the dish. The heart muscle cells condense around these cantilevers to produce tiny miniature beating heart tissues that contract around the micropillar; every time the tissue contracts, the micropillar within it deflects.

Porello said that the device enables researchers to measure the force that the tissues are generating, allowing them to observe how fast the tissues are beating and whether they display any irregularities in their heartbeat. These capabilities are useful for treatment testing because the effect that medication or genetic manipulations of stem cells have on the tissues heartbeat can be seen.

And so it serves as a pretty powerful platform for looking at drug responses, but also modelling genetic forms of heart disease.

Were actually now scaling up these tissues and growing very, very large bioengineered heart tissue patches that can be implanted onto the heart.

In an email to The Epoch Times, Porello said in the future that bioengineered heart tissue patches could be used to treat adults with heart failure, and alternative approaches are already being trialled.

Our bioengineered heart tissues could also be used to support the failing heart in adults with underlying heart disease.

Further studies are required to confirm that our bioengineered heart tissue patches are safe and effective in animal models before progressing to human trials. These pre-clinical safety and efficacy studies are underway.

He noted that although significant advances and a better understanding of the hearts regenerative mechanisms have been made in recent years, using this knowledge to develop a safe and effective drug is a slow process.

It typically takes 10 years and around $1 billion dollars to develop a new heart failure drug and take it all the way through to clinical approval. We are at the beginning of that journey.

We need to gain a better understanding of the fundamental biology underlying heart regeneration before we can develop effective treatments.

Porello is now applying his discoveries in a clinical context at theMCRIto reach his goal of regenerating human hearts. The regeneration research at the institute has two branches, the first focuses on studying diseases using lab-grown models of the heart muscle. The models are made using blood and tissue samples collected from sick children at the Royal Childrens Hospital in Melbourne.

He said that this branch of the research enables the team to model the genetic basis of the disease in any individual.

Were using this technology to model childhood heart disease, trying to understand its causes, and then using those genetic models of heart disease to test and develop therapeutic approaches to treat those conditions, he said.

Porello said that the second branch of the research performed at the MCRI explores the regenerative approach to growing the very, very large bioengineered heart tissue patches. The researchers plan is to eventuallyimplant the patches into a heart to function as a biological assistance device that supports the function of the heart.

If it works, it would be transformative, Porello said.

Stem cells have been used in medicine for more than fifty years, with the most common stem cell procedure currently beingbone marrow transplantsalso known as hematopoietic stem cell transplantsused to treat patients with blood cancers such asleukemiaand blood disorders such assickle cell diseaseandthalassemia.

More recently, skin grown from stem cells has been used to treat extensive burns, and stem cells from fat (adipose tissue) have been used as tissue fillers.

Visit link:
Regenerative Properties of the Newborn Heart Offers Hope for Those With Congenital Heart Disease - The Epoch Times

Posted in Stem Cells | Comments Off on Regenerative Properties of the Newborn Heart Offers Hope for Those With Congenital Heart Disease – The Epoch Times

Page 7«..6789..2030..»