Category Archives: Stem Cell Treatment


Rheumatoid Arthritis Stem Cell Therapy Market Assessment, With Major Top Companies Analysis, Geographic Analysis, Growing Opportunities Data By…

Prophecy Market Research delivered a business report on the Rheumatoid Arthritis Stem Cell Therapy which is the best creation of trust and skill. The report is a top to bottom assessment of the different attributes and future development possibilities during the figure time frame. To uncover every doable way, our examiners applied different strategies. It contains every one of the overall significant organizations to help our clients in understanding their thorough strategies and cutthroat climate.

The noticeable players in the worldwide Rheumatoid Arthritis Stem Cell Therapy are

Mesoblast Ltd., Roslin Cells, Regeneus Ltd, ReNeuron Group plc, International Stem Cell Corporation, TiGenix and others

Our investigator have partitioned the report into segments so you might become familiar with the overall market undiscovered possibility in every one.

Get Sample Copy of This Report @ https://www.prophecymarketinsights.com/market_insight/Insight/request-sample/4815

The market elements are analyzed inside and out in the outline segment. This part is an unquestionable requirement perused for anybody settling on information driven choices. It talks about how Rheumatoid Arthritis Stem Cell Therapy functions, as well as market size and volume. The report is coordinated in straightforward organizations and incorporates outlines, tables, and charts to inspect the information and uncover the secret example in the numbers. Besides, the report incorporates verifiable deals and income data as well as guage designs for the following not many years.

The development and limiting elements are given their own fragment to help our clients in observing the Rheumatoid Arthritis Stem Cell Therapy touch spots and problem areas. The ends attracted this part depend on trustworthy and more significant position sources. Our specialists have utilized an assortment of market projection ways to deal with furnish our clients with reliable outcomes.

The Rheumatoid Arthritis Stem Cell Therapy is isolated into different groupings in the division segment. The fragment is an inside and out assessment of every classification, which is grouped by its qualities and expansiveness. Weve recorded every one of the measurements along with subjective clarifications to assist clients with appreciating the expected broadness of each class before very long. To dispose of errors in current realities and discoveries, the report utilizes an assortment of measurable methodologies. Moreover, an assortment of pattern projection approaches are utilized to uncover future development angles and prospects.

By Product Type (Allogeneic Mesenchymal Stem Cells, Bone Marrow Transplant and Adipose Tissue Stem Cells)

By End-User (Hospitals, Ambulatory Surgical Centers and Specialty Clinics)

By Region (North America, Europe, Asia Pacific, Latin America, and Middle East & Africa)

Mesoblast Ltd., Roslin Cells, Regeneus Ltd, ReNeuron Group plc, International Stem Cell Corporation, TiGenix and others

Promising Regions & Countries Mentioned In The Rheumatoid Arthritis Stem Cell Therapy Report:

The local review area inspects all potential market scenes in specific areas before very long. Its an exhaustive assessment of the Rheumatoid Arthritis Stem Cell Therapy possible districts. The examination additionally remembers a contextual investigation for significant market members to help shoppers distinguish and understand powerful techniques in the overall Rheumatoid Arthritis Stem Cell Therapy , as well as likely boundaries. Our master experts checked the data and endeavored to protect the most ideal degree of exactness.

Segmentation Overview:

By Product Type (Allogeneic Mesenchymal Stem Cells, Bone Marrow Transplant and Adipose Tissue Stem Cells)

By End-User (Hospitals, Ambulatory Surgical Centers and Specialty Clinics)

By Region (North America, Europe, Asia Pacific, Latin America, and Middle East & Africa)

Download PDF Brochure for report overview @ https://www.prophecymarketinsights.com/market_insight/Insight/request-pdf/4815

Purposes Behind Buying Rheumatoid Arthritis Stem Cell Therapy Report-

openings both developed and arising regions.

Understanding the tactics that support marketable interest in terms of products, segmentation, and assiduity verticals can help you make better opinions.

Get In-depth TOC @ https://www.prophecymarketinsights.com/market_insight/Global-Rheumatoid-Arthritis-Stem-Cell-Therapy-Market-4815

Why Buy From Us?

Prophecy Market Insights is particular statistical surveying, investigation, promoting/business technique, and arrangements that offers vital and strategic help to clients for settling on very much informed business choices and to distinguish and accomplish high-esteem valuable open doors in the objective business region. We likewise assist our clients with tending to business challenges and give the most ideal answers for conquer them and change their business.

Contact Us:

Mr. Alex (Sales Manager)

Prophecy Market Insights

Phone: +1 860 531 2701

Email: [emailprotected]

Go here to see the original:
Rheumatoid Arthritis Stem Cell Therapy Market Assessment, With Major Top Companies Analysis, Geographic Analysis, Growing Opportunities Data By...

Sana On Track to Remove Barriers for Cell & Gene Therapy – BioSpace

A Sana scientist works in the lab/Photo courtesy of Sana Biotechnology

One of the most important transformations of 21st-centurymedicine will be the ability to reengineer cells as medicines. Before cell and gene therapy can truly become mainstream medicine, the challenges of allogeneic rejection and targeted delivery must be solved. Sana Biotechnology is on the path to do that, with notable strides that may, just possibly, change medicine as we know it.

Our aspirations are to be able to repair or control genes and gene expression of every cell type in the body thats broken, and to transplant or replace whats missing or too far damaged, Steve Harr, M.D., Sanas president and CEO, told BioSpace.

The scope of that mission is huge. The breadth of our aspirations set Sana apart from most biotech companies, he said. And, Sana is approaching the challenge like any other great endeavor: one step at a time. In this case, that means with ex vivo and in vivo cell engineering platforms.

Sanas ex vivo hypoimmune platform (HIP) is designed to prevent allogeneic rejection, in which the immune system recognizes modified cells as foreign and attacks them. This is perhaps the greatest issue that has held cell therapy back from reaching its potential for patients, Harr said.

The hypoimmune platform appears to solve that hurdle, he said. We have shown in preclinical models that we have the potential to prevent allogeneic rejection.

As Sonja Schrepfer,M.D., Ph.D., head of Hypoimmune Platform, elaborated, The human leukocyte antigens (HLA) on a cells surface are like fingerprints for proteins on the cell. They tell the immune system what is inside your cell. We recognized HLA as a major hurdle (in overcoming rejection) and so prevented expression of the HLA. Now the cells fingerprint is missing.

Of course, viruses figured out long ago that removing HLA can hide cells, so the immune system evolved to be smarter, she continued. NK cells and macrophages recognize cells without HLA (missing self) and attack them. To protect those cells from this attack, we overexpress CD47, which basically is a dont attack me molecule. It protects these cells from being recognized by NK cells and macrophages, allowing them to survive.

These cellular modifications can be performed on pluripotent stem cells, which can then be differentiated into essentially any hypoimmune cell, and also on allogeneic donor cells. We intend to derive hypoimmune pancreatic islet cells to treat type 1 diabetes, for example. Our goal is to provide off-the-shelf cells to anyone, anywhere, at any time that can function without requiring the patient to take immunosuppressive medications, Schrepfer said. Sana also is working to develop T cells, glial progenitor cells and cardiomyocytes as allogeneic therapies.

In preclinical models, We have seen that without immunosuppression, the HIP-edited induced pluripotent stem cells (iPSCs) survived for many weeks without activating the immune system or being rejected, Schrepfer said. That was very exciting! The company is planning to file its first IND to test this system in patients as early as this year.

Targeted gene delivery is the second big challenge Sana is addressing. With antiviral vectors (AAVs), you cant control where the vectors go and, because the cellular DNA is not changed, they have less utility in dividing cells. Lipid nanoparticles (LNPs), another popular delivery vehicle, at this time mainly go to the liver and deliver only RNA and proteins, not DNA, Harr said.

Sanas fusogen platform has the potential to overcome those limitations with in vivo cell and gene modification. You can make just about any modification to genes and gene expression in a petri dish, Harr pointed out, but it is very difficult to deliver the gene modification payload in vivo. Fusogens offer a unique delivery capability for gene editing and gene modification machinery. We have a number of programs in preclinical development, including four that modify T cells for multiple oncology indications, one that modifies hepatocytes to address liver-related disorders and one that modifies hematopoietic stem cells to address hemoglobinopathies.

Sanas science has the potential to disrupt the cell and gene therapy sector of the industry, but its effects on the practice of medicine are still at least several years away. As Harr said, Our key competitive advantage, as a smaller company, needs to be decision-making, which is possible because of greater focus and having the right people.

Harr defines the right people as program heads who are global leaders in their fields. Thats really important, Harr explained, because this isnt as easy as normal drug development (which, itself, isnt easy). For example, Richard Mulligan, Ph.D., head of SanaX, is instrumental in Sanas work to deliver genomic material. SanaX is a distinct research arm within Sana focused around long-term, disruptive improvements in cell and gene therapy. Mulligan was among the first to discover how to insert genes into cells in the 1980s. Terry Fry, M.D., SVP and head of T Cell Therapeutics, is a renowned expert in chimeric antigen receptor T cell (CAR T) therapies and was critical in the development of a number of CAR T cell therapies to date, including Yescarta. Chuck Murry, M.D., Ph.D., SVP and head of cardiometabolic cell therapy, pioneered the use of human pluripotent stem cells for heart regeneration.

The executive leaders also bring a wealth of experience. Some, including Harr, joined Sana after senior positions at Juno Therapeutics. Others honed their skills at Genentech, Amazon, Amgen, Sangamo and the U.S. Food and Drug Administration, amongst others.

Sanas staff of approximately 400 is divided among its facilities in South San Francisco, Seattle and Cambridge, Massachusetts.

The company is expanding, so there are opportunities throughout Sana both geographically and in terms of business and technical specialties. Harr said one hiring priority is in the area of process and analytical development for manufacturing. Another area includes hiring experts in analytic genomics and computational biology to enable in-depth interrogation of the genome, both when youre inserting things into the cells and as stem cells divide and differentiate, he added.

Our goal is to make important medicines that matter for patients, and these roles are essential in doing that. That means the Sana team must also be flexible and resilient.

The world is constantly changing, and we live on the tip of the innovation spear, Harr said. We need people who are intellectually curious and who have grit. Being a pioneer is hard. We will have great days, and we also will have setbacks. We need people who are resilient and who can collaborate effectively on teams. The things we are doing are complex enough that none of us have all the answers. It always takes some of us.

Harr said Sana is committed to ensuring we do all we can to increase individuals chances to succeed and rewarding them fairly. Biotech tries to help people from all over the world. To do that, we need an environment where people can thrive.

Harr continued, Inclusion is the soil from which all great cultures grow. To truly feel included, you need diversity. It helps to know that there are people just like me whove succeeded here. Succeeding is easier when you have that example.

With that philosophy, Sanas Inclusion, Diversity and Equity program is robust. The measure of a great culture is whether it helps you succeed or holds you back. Our measure is to help you be your best self, your true self and to accept people for who they are. This is an exciting time to be at Sana, Harr said.

Sana is transforming from a research company to a research and development company. The movement into development is an important next step, Harr said. Its the chance to prove things work.

Sana is working towards developing important cell and gene therapies for patients. As Harr said, This is a dynamic company with complex science. We have chosen to be audacious.

Featured Jobs on BioSpace

See original here:
Sana On Track to Remove Barriers for Cell & Gene Therapy - BioSpace

Dr. Dietrich on the Current Treatment Landscape of MCL – OncLive

Martin Dietrich, MD, PhD, discusses the current treatment landscape of mantle cell lymphoma.

Martin Dietrich, MD, PhD, physician, Florida Cancer Specialists and Research Institute, assistant professor of internal medicine, University of Central Florida, discusses the current treatment landscape of mantle cell lymphoma (MCL).

Treatment options have drastically evolved in recent years for MCL, which remains an aggressive and complex subtype of lymphoma, Dietrich says. Disease risk assessment and a patients performance status help inform the optimal first-line treatment, Dietrich adds. First-line treatment options include combination chemotherapy and rituximab (Rituxan) for in clinically fit and higher risk patients, and de-escalation chemotherapy strategies for patients with low-risk disease or a poor performance status, Dietrich explains.

Autologous stem cell transplant (ASCT) has carved out a role in the treatment of patients with high-risk disease. However, it is unclear how impactful ASCT will remain in the future with the emergence of better subsequent therapies, Dietrich continues. For now, ASCT is still recommended for many patients, along with maintenance rituximab for patients after initial induction chemotherapy, Dietrich concludes.

See the article here:
Dr. Dietrich on the Current Treatment Landscape of MCL - OncLive

Are COVID-19-Linked Arrhythmias Caused by Viral Damage to the Heart’s Pacemaker Cells? – Weill Cornell Medicine Newsroom

The SARS-CoV-2 virus can infect specialized pacemaker cells that maintain the hearts rhythmic beat, setting off a self-destruction process within the cells, according to a preclinical study co-led by researchers at Weill Cornell Medicine, NewYork-Presbyterian and NYU Grossman School of Medicine. The findings offer a possible explanation for the heart arrhythmias that are commonly observed in patients with SARS-CoV-2 infection.

In the study, reported March 8 in Circulation Research, the researchers used an animal model as well as human stem cell-derived pacemaker cells to show that SARS-CoV-2 can readily infect pacemaker cells and trigger a process called ferroptosis, in which the cells self-destruct but also produce reactive oxygen molecules that can impact nearby cells.

This is a surprising and apparently unique vulnerability of these cellswe looked at a variety of other human cell types that can be infected by SARS-CoV-2, including even heart muscle cells, but found signs of ferroptosis only in the pacemaker cells, said study co-senior author Dr. Shuibing Chen, the Kilts Family Professor of Surgery and a professor of chemical biology in surgery and of chemical biology in biochemistry at Weill Cornell Medicine.

Arrhythmias including too-quick (tachycardia) and too-slow (bradycardia) heart rhythms have been noted among many COVID-19 patients, and multiple studies have linked these abnormal rhythms to worse COVID-19 outcomes. How SARS-CoV-2 infection could cause such arrhythmias has been unclear, though.

In the new study, the researchers, including co-senior author Dr. Benjamin tenOever of NYU Grossman School of Medicine, examined golden hamstersone of the only lab animals that reliably develops COVID-19-like signs from SARS-CoV-2 infectionand found evidence that following nasal exposure the virus can infect the cells of the natural cardiac pacemaker unit, known as the sinoatrial node.

To study SARS-CoV-2s effects on pacemaker cells in more detail and with human cells, the researchers used advanced stem cell techniques to induce human embryonic stem cells to mature into cells closely resembling sinoatrial node cells. They showed that these induced human pacemaker cells express the receptor ACE2 and other factors SARS-CoV-2 uses to get into cells and are readily infected by SARS-CoV-2. The researchers also observed large increases in inflammatory immune gene activity in the infected cells.

The teams most surprising finding, however, was that the pacemaker cells, in response to the stress of infection, showed clear signs of a cellular self-destruct process called ferroptosis, which involves accumulation of iron and the runaway production of cell-destroying reactive oxygen molecules. The scientists were able to reverse these signs in the cells using compounds that are known to bind iron and inhibit ferroptosis.

This finding suggests that some of the cardiac arrhythmias detected in COVID-19 patients could be caused by ferroptosis damage to the sinoatrial node, said co-senior author Dr. Robert Schwartz, an associate professor of medicine in the Division of Gastroenterology and Hepatology at Weill Cornell Medicine and a hepatologist at NewYork-Presbyterian/Weill Cornell Medical Center.

Although in principle COVID-19 patients could be treated with ferroptosis inhibitors specifically to protect sinoatrial node cells, antiviral drugs that block the effects of SARS-CoV-2 infection in all cell types would be preferable, the researchers said.

The researchers plan to continue to use their cell and animal models to investigate sinoatrial node damage in COVID-19and beyond.

There are other human sinoatrial arrhythmia syndromes we could model with our platform, said co-senior author Dr. Todd Evans, the Peter I. Pressman M.D. Professor of Surgery and associate dean for research at Weill Cornell Medicine. And, although physicians currently can use an artificial electronic pacemaker to replace the function of a damaged sinoatrial node, theres the potential here to use sinoatrial cells such as weve developed as an alternative, cell-based pacemaker therapy.

Many Weill Cornell Medicine physicians and scientists maintain relationships and collaborate with external organizations to foster scientific innovation and provide expert guidance. The institution makes these disclosurespublic to ensure transparency. For this information, see profiles for Dr. Todd Evans, and Dr. Robert Schwartz.

Read more here:
Are COVID-19-Linked Arrhythmias Caused by Viral Damage to the Heart's Pacemaker Cells? - Weill Cornell Medicine Newsroom

Stem Cells Turn Into Bone When Sound Waves Are Near – TechTheLead

Share

Share

Share

Email

A breakthrough made by Australian researchers might change the way doctors treat a broken or missing bone. Turns out stem cells can turn into bone if certain conditions are met.

Normally, bone can be made only of mesenchymal stem cells (MSCs) which are biologically found in the bone marrow.

Extracting them from there is a difficult and painful procedure while doing so at scale is beyond tricky.

But this could change any moment now after Australian researchers found that stem cells can be converted into bone when a certain type of sound waves are used.

Tests had previously shown that low frequency vibrations were great at inducing cell differentiation but the process took over a week and the results were mixed at best.

Nobody had bothered to look into high frequency sound waves until now. RMIT researchers took a microchip capable of dispersing sound waves in the Mhz range and turned it at MSCs in silicon oil on a culture plate.

The team noticed that after exposing the cells to 10MhZ signals for 10 minutes daily for five days, the markers indicating the bone conversion appeared.

We can use the sound waves to apply just the right amount of pressure in the right places to the stem cells, to trigger the change process, said Leslie Yeo, co-lead researcher on the study. Our device is cheap and simple to use, so could easily be upscaled for treating large numbers of cells simultaneously vital for effective tissue engineering.

This discovery, detailed in the journalSmall, eliminates the need of drugs to make stem cells behave this way. Moreover, the MSCs can be pulled from a variety of places, like fast tissue, not just bone marrow.

By injecting them into the body in case of an injury or disease, they can start working on a new bone faster and more efficient.

Watch These Goldfish Drive a Wheeled Platform!

Paralyzed Man Sends Tweet With Brain Chip In a Worlds First

Facebook Twitter LinkedIn Reddit WhatsApp

Subscribe to our website and stay in touch with the latest news in technology.

You will soon receive relevant content about the latest innovations in tech.

There was an error trying to subscribe to the newsletter. Please try again later.

See the rest here:
Stem Cells Turn Into Bone When Sound Waves Are Near - TechTheLead

Maryland Cancer Moonshot Initiative Promises $216 Million for Research and Treatment Conduit Street – Conduit Street

Maryland will be committing $216 million to expand and accelerate cancer detection, screening, prevention, treatment, and research through Governor Larry Hogans recently announced Maryland Cancer Moonshot Initiative.

Governor Larry Hogan explained the personal significance of the initiative in a press release:

The reality is that cancer is a disease that has touched nearly every one of us, through family or loved ones, saidGovernor Hogan. On the day I found out I was cancer-free, I pledged that as long as I am governor and long after, I will stand with all those who are fighting this terrible disease. That is why today, I am announcing the Maryland Cancer Moonshot, to dramatically accelerate all of our efforts to detect, prevent, treat, and find a cure for cancer, so that more lives can be saved. This is a watershed moment in the fight against cancer in our state and the region.

The substantial initial investment is a part of Governor Hogans fifth supplemental budget and will include funding for the following:

Greenebaum Cancer Center:$100 million for the expansion of the University of Maryland Medical Systems Greenebaum Comprehensive Cancer Center (UMGCCC) in downtown Baltimore to providestate-of-the-art inpatient and outpatient cancer services. UMGCCC, which is a National Cancer Institute-designated comprehensive cancer center, treats approximately 3,000 new patients annually. This investment completes the states commitment to the project.

Prince Georges Comprehensive Cancer Center:$67 million to fully fund the construction of a new comprehensive cancer center on the campus of the newUniversity of Maryland Capitol Region Medical Centerin Largo. This best-in-class cancer will be a premiere clinical and research center to serve the residents of Prince Georges County and the region. The state funding includes a $27 million commitment by the governor, a $13.5 million commitment by the Maryland Senate and a $26.5 million commitment by the Maryland House of Delegates.

Cancer Research:$25 million for the University of Maryland School of Medicine and Johns Hopkins University to accelerate cancer research projects.

Pediatric Cancer Research:$1 million to support expanding pediatric cancer research at the University of Maryland School of Medicine.

Stem Cell Research Fund:$20.5 million for the Maryland Stem Cell Research Fund (MSCRF) to catalyze investment in regenerative medicine projects to develop novel cures and groundbreaking treatments for prevalent cancers.

Maryland Tech Council:$2.5 million for the BioHub Maryland Initiative to expand the states life sciences and biotechnology research workforce, with a focus on talent development, upskilling opportunities, and outreach to students in underserved communities. Maryland is proud to be home to one of thetop biotech clustersin the United States.

Read the full press release.

Like Loading...

Read the original here:
Maryland Cancer Moonshot Initiative Promises $216 Million for Research and Treatment Conduit Street - Conduit Street

Orchard Therapeutics Extends Runway into 2024, Focusing HSC Gene Therapy Platform Exclusively on Severe Neurometabolic Diseases and Research Platform…

Libmeldy European launch momentum building with multiple MLD patients treated and strong recognition of value proposition; U.S. BLA filing of OTL-200 on track for late 2022 / early 2023

Broad research platform presents opportunities for larger indications and partnerships; preclinical POC data in NOD2 Crohns disease expected by year end and IND filing planned in 2024

Plan to seek strategic alternatives for primary immunodeficiency programs, including OTL-103 in WAS

Refined portfolio and 30% proposed workforce reduction extend cash runway into 2024

BOSTON and LONDON, March 30, 2022 (GLOBE NEWSWIRE) -- Orchard Therapeutics (Nasdaq: ORTX), a global gene therapy leader, today announced its intention to focus its hematopoietic stem cell (HSC) gene therapy platform exclusively on severe neurometabolic diseases and early research programs while also reporting its financial results for the quarter and year ended December 31, 2021. These actions are intended to extend the companys cash runway into 2024 and focus operations on the highest value programs in its portfolio.

Moving forward, Orchard will continue its investment in Libmeldy (atidarsagene autotemcel) / OTL-200 for metachromatic leukodystrophy (MLD) to help sustain recent commercial momentum in Europe, as well as to support regulatory and future commercial activities for a potential U.S. approval and launch. The company also will continue to advance clinical development of OTL-203 for mucopolysaccharidosis type I Hurlers syndrome (MPS-IH) and OTL-201 for mucopolysaccharidosis type IIIA (MPS-IIIA). The focus on these neurometabolic programs is expected to allow Orchard to leverage the clinical validation of HSC gene therapy demonstrated with Libmeldy and capture significant commercial synergies, especially given the immense unmet needs in these diseases. Promising early-stage research programs that apply the HSC gene therapy approach in NOD2 Crohns disease, hereditary angioedema (HAE) and progranulin mutated frontotemporal dementia (GRN-FTD) also will remain an important part of the portfolio going forward given their promise in larger indications and as a possible source of future partnerships.

In light of our experiences and knowledge gained in this current and rapidly evolving market environment for gene therapy, our plan is to concentrate resources on programs that have the potential to make a remarkable difference to patients while also providing sustainable value to the business to enable the achievement our long-term vision, said Bobby Gaspar, M.D., Ph.D., chief executive officer. As launch momentum for Libmeldy continues to build in Europe and we prepare for a regulatory filing in the U.S., a focused strategy that utilizes a common infrastructure for future neurometabolic disease launches is critical to our success as a commercial gene therapy company.

Latest Key Milestones

To advance its portfolio of gene therapies for neurometabolic disorders and investigate future applications for the HSC approach, Orchard has provided an updated list of expected milestones:

Libmeldy Recent Highlights

Portfolio and Organizational Updates

To support the companys refined strategic focus and provide runway extension into 2024, Orchard intends to discontinue its investment in and seek alternatives for its programs in rare primary immune deficiencies. These include OTL-103 for the treatment of Wiskott-Aldrich syndrome (WAS), OTL-102 for X-linked chronic granulomatous disease (X-CGD) and Strimvelis, a gammaretroviral vector-based gene therapy approved in Europe for adenosine deaminase severe combined immunodeficiency (ADA-SCID).

Regarding the regulatory status of the OTL-103 program in the U.S., Orchard recently received written feedback from the FDA. The company believes the path to a potential BLA filing may require additional time and further investment.

Gaspar continued, We recognize the significant need that persists for many patients suffering from these rare diseases of the immune system, and we sympathize with the individuals, families and healthcare providers affected by these announcements, as well as our clinical partners and colleagues who worked so hard to advance these programs. These therapies have shown the potential for significant benefit for many patients treated in the clinical studies and we will continue to look for alternative ways to advance them, which could include commercial partnerships.

As a result of these updates, the company has proposed to reduce its current workforce by approximately 30%, which will result in a restructuring charge in 2022. Collectively, the actions announced today are expected to extend the companys existing cash runway into 2024.

Fourth Quarter 2021 Financial Results

Research and development expenses were $23.3 million for the three months ended December 31, 2021, compared to $22.6 million in the same period in 2020. R&D expenses include the costs of clinical trials and preclinical work on the companys portfolio of investigational gene therapies, as well as costs related to regulatory, manufacturing, license fees and milestone payments under the companys agreements with third parties, and personnel costs to support these activities. The company expects R&D expenses to decline beginning in the second quarter of 2022 due to the portfolio updates and workforce reduction announced today as well as the completion of activities to support the OTL-200 BLA submission.

Selling, general and administrative expenses were $13.6 million for the three months ended December 31, 2021, compared to $16.2 million in the same period in 2020. The decline from 2020 resulted primarily from lower cash and share-based personnel costs to align with the current filing timelines and commercialization plans. In 2022, the company expects SG&A expenses to decline from 2021 due to the workforce reduction announced today, partially offset by increasing commercialization expenses to support Libmeldy, including preparations for a potential U.S. launch in 2023.

Net loss was $36.4 million for the three months ended December 31, 2021, compared to $ 33.6 million in the same period in 2020. The company had approximately 125.7 million ordinary shares outstanding as of December 31, 2021.

Cash, cash equivalents and investments as of December 31, 2021, were approximately $220.1 million, with $33.0 million of debt outstanding, compared to $191.9 million and $25.0 million of debt outstanding as of December 31, 2020. Following the actions announced today, the company now expects that its existing cash, cash equivalents and investments will fund its anticipated operating and capital expenditure requirements into 2024.

Conference Call & Webcast Information

Orchard will host a conference call and live webcast with slides today at 8:00 a.m. ET to discuss the updates to its business strategy. The conference call will be broadcast live in listen-only mode under "News & Events" in the Investors & Media section of the company's website at http://www.orchard-tx.com, and a replay will be archived on the Orchard website following the presentation. To ask a question, please dial (866) 987-6504 (U.S. domestic) or +1 (602) 563-8620 (international) and refer to conference ID 9445456. Please dial in at least 15 minutes in advance to ensure a timely connection to the call.

About Libmeldy / OTL-200

Libmeldy (atidarsagene autotemcel), also known as OTL-200, has been approved by the European Commission for the treatment of MLD in eligible early-onset patients characterized by biallelic mutations in the ARSA gene leading to a reduction of the ARSA enzymatic activity in children with i) late infantile or early juvenile forms, without clinical manifestations of the disease, or ii) the early juvenile form, with early clinical manifestations of the disease, who still have the ability to walk independently and before the onset of cognitive decline. Libmeldy is the first therapy approved for eligible patients with early-onset MLD.

The most common adverse reaction attributed to treatment with Libmeldy was the occurrence of anti-ARSA antibodies. In addition to the risks associated with the gene therapy, treatment with Libmeldy is preceded by other medical interventions, namely bone marrow harvest or peripheral blood mobilization and apheresis, followed by myeloablative conditioning, which carry their own risks. During the clinical studies of Libmeldy, the safety profiles of these interventions were consistent with their known safety and tolerability.

For more information about Libmeldy, please see the Summary of Product Characteristics (SmPC) available on the EMA website.

Libmeldy is approved in the European Union, UK, Iceland, Liechtenstein and Norway. OTL-200 is an investigational therapy in the U.S.

Libmeldy was developed in partnership with the San Raffaele-Telethon Institute for Gene Therapy (SR-Tiget) in Milan, Italy.

About Orchards Investigational Primary Immune Deficiency Portfolio

Primary immune deficiencies (PIDs) are a group of rare, genetic disorders in which the immune system does not function properly, leading to frequent infections and other disease manifestations that can be life-threatening. Orchards PID portfolio includes HSC gene therapies in development for the treatment of Wiskott Aldrich syndrome (WAS), X-linked chronic granulomatous disease (X-CGD), and Adenosine deaminase severe combined immunodeficiency (ADA-SCID). More than 100 PID patients have received one of Orchards investigational gene therapy products, with 11 years follow-up in the earliest treated patients. The majority of patients experienced favorable clinical outcomes and there was no evidence of monoclonal expansion, leukoproliferative complications or emergence of replication competent lentivirus.

About Strimvelis

Strimvelis(autologous CD34+enriched cell fraction that contains CD34+cells transduced with retroviral vector that encodes for the human ADA cDNA sequence)is a gammaretroviral vector-based gene therapy approved by the European Medicines Agency (EMA) in 2016. It was the firstex vivoautologous gene therapy approved by the EMA. Strimvelishas not been approved by the U.S. Food and Drug Administration (FDA).

Strimvelis is indicated for the treatment of patients with severe combined immunodeficiency due to adenosine deaminase deficiency (ADA-SCID), for whom no suitable human leukocyte antigen (HLA)- matched related stem cell donor is available. Strimvelis is intended solely for autologous use and must be given in a specialized hospital by a doctor who is experienced in treating patients with ADA-SCID and in using this type of medicine.

Serious adverse reactions include autoimmunity (e.g., autoimmune hemolytic anemia, autoimmune aplastic anemia, autoimmune hepatitis, autoimmune thrombocytopenia and Guillain-Barr syndrome). The most commonly reported adverse reaction was pyrexia.

For more information about Strimvelis, please see the EU Summary of Product Characteristics available on theEMA website.

About Orchard Therapeutics

At Orchard Therapeutics, our vision is to end the devastation caused by genetic and other severe diseases. We aim to do this by discovering, developing and commercializing new treatments that tap into the curative potential of hematopoietic stem cell (HSC) gene therapy. In this approach, a patients own blood stem cells are genetically modified outside of the body and then reinserted, with the goal of correcting the underlying cause of disease in a single treatment.

In 2018, the company acquired GSKs rare disease gene therapy portfolio, which originated from a pioneering collaboration between GSK and the San Raffaele Telethon Institute for Gene Therapy in Milan, Italy. Today, Orchard is advancing a pipeline spanning pre-clinical, clinical and commercial stage HSC gene therapies designed to address serious diseases where the burden is immense for patients, families and society and current treatment options are limited or do not exist.

Orchard has its global headquarters in London and U.S. headquarters in Boston. For more information, please visitwww.orchard-tx.com, and follow us onTwitterandLinkedIn.

Availability of Other Information About Orchard Therapeutics

Investors and others should note that Orchard communicates with its investors and the public using the company website (www.orchard-tx.com), the investor relations website (ir.orchard-tx.com), and on social media (twitter.com/orchard_tx and http://www.linkedin.com/company/orchard-therapeutics), including but not limited to investor presentations and investor fact sheets, U.S. Securities and Exchange Commission filings, press releases, public conference calls and webcasts. The information that Orchard posts on these channels and websites could be deemed to be material information. As a result, Orchard encourages investors, the media, and others interested in Orchard to review the information that is posted on these channels, including the investor relations website, on a regular basis. This list of channels may be updated from time to time on Orchards investor relations website and may include additional social media channels. The contents of Orchards website or these channels, or any other website that may be accessed from its website or these channels, shall not be deemed incorporated by reference in any filing under the Securities Act of 1933.

Forward-looking Statements

This press release contains certain forward-looking statements about Orchards strategy, future plans and prospects, which are made pursuant to the safe harbor provisions of the Private Securities Litigation Reform Act of 1995. Such forward-looking statements may be identified by words such as anticipates, believes, expects, plans, intends, projects, and future or similar expressions that are intended to identify forward-looking statements. Forward-looking statements include express or implied statements relating to, among other things, Orchards business strategy and goals, the therapeutic potential of Orchards products and product candidates, including the products and product candidates referred to in this release, Orchards expectations regarding the timing of regulatory submissions for approval of its product candidates, including the product candidates referred to in this release, the timing of interactions with regulators and regulatory submissions related to ongoing and new clinical trials for its product candidates, the timing of announcement of clinical data for its product candidates, the likelihood that such data will be positive and support further clinical development and regulatory approval of these product candidates, the likelihood of approval of such product candidates by the applicable regulatory authorities, the size of the potential markets for Libmeldy and Orchards other product candidates, the expected benefits to Orchards business as a result of the organizational updates referred to in this release, the adequacy of the companys manufacturing capacity and plans for future investment, and the companys financial condition and cash runway into 2024. These statements are neither promises nor guarantees and are subject to a variety of risks and uncertainties, many of which are beyond Orchards control, which could cause actual results to differ materially from those contemplated in these forward-looking statements. In particular, these risks and uncertainties include, without limitation: that the cost of discontinuing or partnering programs may be higher than expected; the risk that Orchard will not realize the anticipated benefits of its new strategic plan or the expected cash savings; the risk that any one or more of Orchards product candidates, including the product candidates referred to in this release, will not be approved, successfully developed or commercialized; the risk of cessation or delay of any of Orchards ongoing or planned clinical trials; the risk that Orchard may not successfully recruit or enroll a sufficient number of patients for its clinical trials; the risk that prior results, such as signals of safety, activity or durability of effect, observed from preclinical studies or clinical trials will not be replicated or will not continue in ongoing or future studies or trials involving Orchards product candidates; the delay of any of Orchards regulatory submissions; the failure to obtain marketing approval from the applicable regulatory authorities for any of Orchards product candidates or the receipt of restricted marketing approvals; the risk of delays in Orchards ability to commercialize its product candidates, if approved; the risk that the ongoing and evolving COVID-19 pandemic could affect the company's business; and the risk that the market opportunity for Libmeldy and its other product candidates may be lower than estimated. Given these uncertainties, the reader is advised not to place any undue reliance on such forward-looking statements.

Other risks and uncertainties faced by Orchard include those identified under the heading "Risk Factors" in Orchards most recent annual or quarterly report filed with the U.S. Securities and Exchange Commission (SEC), as well as subsequent filings and reports filed with the SEC. The forward-looking statements contained in this press release reflect Orchards views as of the date hereof, and Orchard does not assume and specifically disclaims any obligation to publicly update or revise any forward-looking statements, whether as a result of new information, future events or otherwise, except as may be required by law.

Condensed Consolidated Statements of Operations Data (In thousands, except share and per share data) (Unaudited)

Condensed Consolidated Balance Sheet Data (in thousands) (Unaudited)

Contacts

Investors Renee Leck Director, Investor Relations +1 862-242-0764 Renee.Leck@orchard-tx.com

Media Benjamin Navon Director, Corporate Communications +1 857-248-9454 Benjamin.Navon@orchard-tx.com

Read the original post:
Orchard Therapeutics Extends Runway into 2024, Focusing HSC Gene Therapy Platform Exclusively on Severe Neurometabolic Diseases and Research Platform...

Role of Stem-Cell Transplantation in Leukemia Treatment

Stem Cells Cloning. 2020; 13: 6777.

1Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia

1Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia

1Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia

2Department of Immunology and Molecular Biology, School of Biomedical and Laboratory, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia

1Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia

2Department of Immunology and Molecular Biology, School of Biomedical and Laboratory, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia

Correspondence: Gashaw Dessie Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia, Phone: Tel +251-97-515-2796, Email dessiegashaw@yahoo.com

Received 2020 May 15; Accepted 2020 Jul 25.

Stem cells (SCs) play a major role in advanced fields of regenerative medicine and other research areas. They are involved in the regeneration of damaged tissue or cells, due to their self-renewal characteristics. Tissue or cells can be damaged through a variety of diseases, including hematologic and nonhematologic malignancies. In regard to this, stem-cell transplantation is a cellular therapeutic approach to restore those impaired cells, tissue, or organs. SCs have a therapeutic potential in the application of stem-cell transplantation. Research has been focused mainly on the application of hematopoietic SCs for transplantation. Cord blood cells and human leukocyte antigenhaploidentical donors are considered optional sources of hematopoietic stemcell transplantation. On the other hand, pluripotent embryonic SCs and induced pluripotent SCs hold promise for advancement of stem-cell transplantation. In addition, nonhematopoietic mesenchymal SCs play their own significant role as a functional bone-marrow niche and in the management of graft-vs-host disease effects during the posttransplantation process. In this review, the role of different types of SCs is presented with regard to their application in SC transplantation. In addition to this, the therapeutic value of autologous and allogeneic hematopoietic stemcell transplantation is assessed with respect to different types of leukemia. Highly advanced and progressive scientific research has focused on the application of stem-cell transplantation on specific leukemia types. We evaluated and compared the therapeutic potential of SC transplantation with various forms of leukemia. This review aimed to focus on the application of SCs in the treatment of leukemia.

Keywords: stem cell, leukemia, transplantation

Stem cells (SCs) are undifferentiated cells that can be differentiated into other types of cell andalso have the potential to proliferate and self-renew to producenew SCs. In mammals, there are two broad type of SC. Embryonic SCs (ESCs) are present in the early life of the embryo and isolated from the inner cell massor morula of the blastocyst (future germ layer, such as endoderm, ectoderm, or mesoderm of the embryo).14 The surrounding section of the morula is known as the trophoblast, which can develop to the future placenta. Adult SCs (ASCs) are found in various tissue types of developed mammals.5 ASCs are useful for tissue regeneration and repair after severe injuries.1,6

SC populations may behave abnormally or be altered by genetic or environmental factor, resulting in the development of cancer. Leukemia comprises a group of hematologic disorders that usually begin in the bone marrow and resultin a high number of abnormal blood cells. It is the result of deregulation of normal hematopoietic SC (HSC) development by genetic mutation that produces a cell population known as leukemic SCs (LSCs). The generation of blood cells depends on the regulation of differentiation and proliferation characteristics of HSCs.7 Deregulated differentiation and proliferation activity of HSCs, including chromosomal translocation and somatic mutation, leads to different hematologic disorders. There are four major abnormalities identified under LSCs: such as acute myeloid leukemia (AML), acute lymphoblastic leukemia (ALL),8 chronic LL (CLL) and chronic ML (CML).4 Leukemia and lymphoma (Hodgkins lymphoma [HL] and non-HL [NHL]) are the two major types of blood cancers that result from uncontrolled proliferation of white blood cells, and were the first to be treated clinically using HSC transplantation (HSCT).1,9-11 In addition, HSCT is used as a therapeutic option for many nonhematopoietic malignancies, aplastic anemia, and certain inherited disorders like severe thalassemia, sickle-cell disease, and other inherited metabolic disorders. Historically, HSCs were obtained only from bone marrow, but are now mostly harvested from peripheral blood after mobilization through administration of hemaTtopoietic growth factor and from the umbilical cord blood (UCB) of newborns.4,9

SC-based therapies become the major concern of researchers after the first effective bone-marrow transplant in 1968.12 Globally, food and drug administrations design regulations on the application of SC therapies. An increase in scientific knowledge of cell-differentiation pathways has promoted the application of SC therapy.12 Since the application of SC therapy emerged as a new insight into cellular therapeutic potential, food and drug administrations have continuously driven awareness and designed regulation with regard to SC therapies. SCs serve as a novel cellular therapeutic approach in the field of regenerative medicine to treat various disorders.13 In addition to renewing and proliferating themselves, they are capable of differentiation to specialized functional cells.14 This enables them to substitute various injured cells, such as cardiomyocytes, fibroblasts, and endothelial cells.15 In addition, regenerative medicine has significant therapeutic potential through the application of SCT to restore impaired blood cells.16

HSCT has broad application in treating different malignant and nonmalignant hematologic disorders. Researchers have noted that >40,000 HSCTs are performed every year to treat these disorders.17 In this context, autologous SCT (auto-SCT) and allogeneic SCT (allo-SCT) are the best known and most applicable.18 There are SC types that have the capability of being the source for SCTs. Bone-marrow SCs are the major sources for treating hematologic and nonhematologic disorders.19 Similarly, peripheral blood CD34+ cell have hematopoiesis potential for HSCT.20 With respect to recent scientific advancement,HSCs are generated from pluripotent ESCs that require the transition state from endothelial to hematopoietic progenitor cells to resolve HLA-mismatched problem.21 The recent investigation done by Serap et al (2019) and his colleagues hypothesized that achievement of effective HSCT may also associate with non-hematopoietic progenitor cells, very small embryonic-like SCs (VSELSCs).22 They differentiate into HSCs in vitro.23 With specific forward reprogramming protocols, induced pluripotent SCs (iPSCs) have therapeutic potential to generate hemato-endothelial progenitor (HEP) cells.

Co-administration of chemotherapy along with auto-SCT leads to a decrease in the level of regulatory T-cells. In response to the dysregulated immune system, biological characteristics of mesenchymal SCs (MSCs) contribute to hematopoietic reconstitution and an efficient HSC engraftment.24,25 On the other hand, bone marrow derived MSCs are other components of hematopoietic niche.26 Therefore, this review assessed different types of SCs that are utilized as the source and as support of SC transplantation. In addition, we also summarized the role of allogeneic and auto-SCT in the treatment of various types of leukemia.

The involvement of ESCs is the new therapeutic insights having a regenerative potential to restore impaired tissue or cells.27 ESCs are the source of SCs for cellular transplantation therapies; however, they may also lead to uncontrolled cell proliferation which also results in the development of cancers.28 The challenges of using these cells are their characteristic features of chromosomal abnormality and mutation during in vitro.29 Regard to this, c-MYC oncogene may be expressed that results in cancer cells than their cellular therapeutic significant.29 They require a safety concern due to their teratoma formation.30 Although they have teratoma effect, ESCs have a significant role in the transplantation process.28 Human ESCs (hESCs) serve as the source of development of cellular lineages through signaling pathways.13 Recently, protocols have been on the way to be designed to generate HSCs from pluripotent ESCs in vitro. The generation of HSCs from those pluripotent ESCs requires a transition from endothelial to hematopoietic progenitor cells to resolve HLAmismatching.21 The hematopoietic transcription factor Runx1 promotes the commitment of hematopoietic cellular lineages by activating the expression of Runx1a. NOTCH signaling enhances the transition state, while the TGF-signaling pathway inhibit it.31 Recently, generation of HSCs was achieved by Wang et al from hESCs andhumaniPSCs (). The commitment stages that had been examined by those scientists confirmed the synthesis of hematopoietic cells from hESCs.32 In support of this, recently the ESC gene SLL4was identified and used as a therapeutic target for leukemia. Because of its importance in the ESC fate, SALL4 expression need to be reactivated during the reprogramming process of mouse embryonic fibroblasts to be converted into iPSCs. Under normal condition, SALL4 is expressed highly in CD34+CD38 HSCs and llittle in CD34+CD38 + hematopoietic progenitor cells. Therefore, the main application behind this ESC gene product is as key player in hematopoietic differentiation. Consequently, downregulation of this gene could be considered a therapeutic option for leukemia.33

Role of different types of SCs in SC transplantation. MSCs were the nonhematopoietic source utilized to reduce GVHD (reduce risk of graft failure by secreting soluble factors with anti-inflammatory properties), efficient HSCs support to engraftment of transplant, hematologic reconstitution, and to improve the HSCT outcome. HSCs can be generated from the hematoendothelial transition process from HESCs to HiPSCs, and commonly from bone-marrow SCs, PBSCs, and umbilical cord blood. The pluripotent potential of VSELSCs also enables to generate HSCs.

Abbreviations: GVHD, graft-vs-host disease; HESCs, human embryonic SCs; HSCs, hematopoietic SCs; HSCT, hematopoietic SC transplantation; HiPSCs, human induced pluripotent SCs; MSCs, mesenchymal SCs; PBSC, peripheral blood SC; VSELSCs, very small embryonic-like SCs.

iPSCs were introduced as an alternative SC-based therapy method in 2006, by Takahashi and Yamanaka.34 Reprogramming of SCs through the integration of viruses with these cells induces differentiation capability in various tissue types.35 These are pSCs, which are generated from adult somatic cells through in vitro experimental investigation.36 They are synthesized in vitro by reprogramming mature mouse fibroblast cells through epigenetic modification.34 In human beings, production of iPSCs was started through the introduction of four genes SOX2, MYC, OCT4, and KLF4 into matured somatic fibroblasts37 and other human somatic cells.38 The genes are induced in these cells through the encoded retrovirus.39 The ability of iPSCs to expand into multicellular lineages enables them to be a potential SC-therapy method. Various types of patient-specific SCs have been synthesized from their expansion process in vitro.40 Research has revealed their cellular therapeutic significance in various hematologic malignancies, such as CML, MDS, AML,22 and BCR-ABLmyeloproliferative neoplasms.41 Donor blood cells are reprogrammed to iPSCs to generate patient-specific SCs.40 With specific forward-reprogramming protocols, iPSCs have the therapeutic potential to generate hematoendothelial progenitor cells. Lange et al demonstrate the possible generation of hematopoietic progenitor cells by combinatorial expression of transcription factors SCL, LMO2, GATA2, and ETV242 (). Moreover, researchers have been trying to generate hematopoietic progenitor cells from PSCs. Shan et al described possible strategies for generation of HSCs from human mesenchymal cells with hematopoietic potential (). They revealed the derivation or generation of hematopoietic progenitor cells from mouse PSCs using in vitro induction methods. Therefore, iPSCs can be have possible therapeutic potential in SCT; however, they present safety concerns, due to their teratoma formation.30 Allogeneic transplantation of bone marrow or umbilical cord reveals rejection, due to the effect of graft-vs-host disease (GVHD) and disease relapse, which restricts its applicability. In cases of auto-HSCT, there is no risk of rejection, but there remain leukemic cells that induce disease relapse. Collectively, these disadvantages of bone-marrow HSCT mandate alternative sources of HSCs aiming to reduce GVHD, disease relapse, and bone marrowfailure syndrome. Considering this, iPSCs represent a suitable source to generate HSCs in vitro with limited immunogenicity.43 These have a major advantage over bone-marrow and cord types, since their autologous transplantation from iPSCs does not induce GVHD.44

Bhartiya et al characterized VSELSCs as the true SCs and the subset of different SC population, such as HSCs, ovarian SCs and MSCs. They express the OCT4A antigenic marker in their nucleus.30 The pluripotency features of VSELSCs enhance their expansion in vitro using the pyrimidoindole-derivative molecule UM171,45 and in turn are utilized for expansion of CD34+ HSCs.46 VSELSCs are involved in homeostatic processes, because they are found in quiescent stage, and later they differentiate into ASCs. They differentiate into HSCs in vitro.23 VSELSCs can be generated from primordial germ cells and undergo further differentiation into HSCs47 (). Bone marrowderived VSELSCs may not have features characteristic of hematopoietic progenitor SCs, but they can retain hematopoietic features through external-stress growth factors.48 The transcriptional factors Oct4A), Nanog, and Rex1 are found in VSELSCs, but they are not expressed in HSCs.22 Treatment of immunocompromised ALL8 patients with granulocyte colonystimulating factorincreases mobilization of VSELSCs to the peripheral circulation.49 Dissemination of VSELSCs to the circulation promotes the regeneration of tissue.49 A recent investigation done by Serap et al hypothesized that achievement of effectiveHSCT may be associated with nonhematopoietic progenitor cells VSELSCs.22 The expression of transcription factors and pluripotent markers may contribute to their therapeutic potential in SC transplantation. Demonstrations on immunocompromised mice have shown that VSELSCs have a lower teratoma effect.47 Similarly, an investigation done on animal models showed that they have the capability to differentiate into HSCs.46

Bone marrowderived MSCs are important to regenerate injured tissue.50 Recently, MSCs have served as a new cellular therapy method in the field of regenerative medicine.13 They inhibit cancer-cell proliferation through secretion and inhibition of Dkk1- and Wnt-signaling pathways, respectively.51 Besides this, MSCs alter the immune system to regenerate damaged tissue and decrease inflammation.52 GVHD is one of the complications of both auto-SCT and allo-SCT during treatment.53 This posttransplantation complication is associated with immunologic intolerance.53 Indeed, MSCs have been shown to support the engraftment of autologously or allogeneically transplanted HSCs by secreting soluble factors or immunomodulators, such as TGF1 and HGF which inhibit the proliferation of CD4+ TH1, TH17, CD8+ T, and natural-killer cells, leading to prevention of GVHD.6,24,26 Therefore, GVHD that occurs after HSCT can be treated by coinfusion with MSCs.54 Bone marrowderived MSCs are components of the hematopoietic niche. Additionally, they have the capability to regulate the hematopoiesis process through interactionand communicating with HSCs and progenitor cells55 ().

Donor availability is a very important issue, particularly in patients from ethnic minorities. A haploidentical donor and CB allow allo-HSCT in the majority of transplant-eligible patients.UCB is a well-established cellular product source for hematopoietic reconstitution and transplantation.37 It is derived from fetal tissue and acts as a potential source of progenitor SCs to synthesize matured HSCs16 (). The lower complication rate of GVHD and less stringent HLA-matching requirements make it a valuable source of HSCs.56 It is more highly enriched with HSCs/progenitor cells than peripheral blood with regard to colony-forming unitgranulocyte/macrophage progenitors and CD34+-cell content.57

The effect of HLA mismatching is less severe in mismatched UCB transplantation than unrelated peripheral and bone marrowblood transplantation;58 therefore, higher numbers of mismatched donors may donate to save lives. Compatibility at the DRB1-allele and HLA-A and -B antigen level is better for UCB transplantation to be selected traditionally without consideration of HLA-C.59 UCB has significance for allo-HSCT transplantation, because it requires lower HLA matching than for unrelated donors.59 In AML, unrelated CB transplantation has failed, due to nonrelapse mortality.60 However, the cost of CB delaying engraftment and risk of infection are still challenges in its application for hematologic diseases, including leukemia.61,62

In cases of rapid requirement of allograft and absence of an HLA-matched donor, HLA-haploidentical SC transplantation is considered a therapeutic option.63 Peripheral and bone-marrow SCs can be donated from these family members if they have one common haplotype.64 HLA-haploidentical cells are considered an optional source for HSCT.65 In haploidentical transplantation, the graft contains lower of T-cell content to diminish GVHD.66 Outcomes of haploidentical HSCT may be affected by innate immune cells like T cells and natural-killer cells.67 In high-risk acute leukemia, the applicabilion of HLA-haploidentical HSCT is elevated.65 However, outcomes of nonrelapse mortality and GVHD may be increased from haploidentical HSCT with higher HLA mismatching including from partially related donors, as the content of T-cell is replete.68

A soft, gelatinous tissue, bone marrow is used as the source of peripheral HSCs.69 Researchers have argued that both bone marrow and peripheral blood are major sources of SCs. SCASCs generated from bone marrow are known as bone-marrow SCs,37 having clinical significance in restoring damaged cardiac tissue through gene therapy.70 Also, they can be a potential source for auto-HSCT..37 There is an improvement in GVHD in patients with bone-marrow SC transplantation compared to peripheral blood SCs (PBSCs).19 Bone marrowSC transplantation is utilized in various hematologic malignancies, such as AML, ALL, and CML. The use of bone-marrow transplantation from compatible donors is the most effective treatment for CML.71 Allogenic bone-marrow transplantation is an effective alternative treatment option for patients who are resistant to chemoradiation therapy and have a higher probability of relapse.72 The physician removes marrow from the donors hip bone using surgical procedures, including anesthesia, sterile needles, and syringes, and replaces the donated bone marrow within 46 weeks. As the level of T cell compare in both bone-marrow transplantation and PBSCs, the concentration of T cells is reduced in bone-marrow transplantation.19

Recent SC-transplantation protocols state that mobilization of HSCs from bone marrow to peripheral blood is an effective treatment method in the majority of transplanted patients.73 Although bone marrow is major source of SCs, a hematopoietic growth factor found in PBSCs showed that these are also another possible source of SCs.74 PBSCs from bone marrow are a valuable source in restoring hematologic disorders.69 The potential effect of PBSCs depends on hematopoietic development and enhancement of immunologic profiles, and hence they are a valuable source of HSCs to treat hematologic disorders. Peripheral blood CD34+ cells have hematopoietic potential for SCT.20 Javarappa et alpurified hematopoietic progenitor cells from CD4+ peripheral blood cellsafter which the cells differentiated into megakaryocytes and myeloid-lineage cells75 (). PBSCs serve as a valuable SC source if mobilization is supported by granulocyte colonystimulating factor.19 They are applicable in autolo-SCT in the treatment of multiple myeloma.76 The utilization of peripheral SCs as a source of SCs may induce the occurrence of GVHD.77 Even if they have such effects, the immune system has been enhanced, due to elevation of T-cell secretion. On the contrary, the elevation of T cells may also cause GVHD development;19 however, PBSC collection in children may expose them to metabolic complications, including hypocalcemia and hypoglycemia.78

The tight control in proliferation and differentiation of HSCs has significant value for the synthesis of blood cells.7 Multipotent HSCs are responsible for cell division and proliferation.79 Somatic mutation of T cells during DNA methylation and posttransplantation alteration are risk factors for ALL.8,80 CML is a hematologic disorder induced by reverse chromosomal translocation on t(9;22)(q34;q11)81 and BCRABL oncogene effects on proliferative myelogenous cells.82 Mutated gene BCRABL, has a tyrosine-kinase effect and induces the release of highly proliferative myelogenous cells from bone marrow.81 The MYC gene is another oncogene that induces gene expression and has a proliferative effect on hematopoietic progenitor cells.83 In addition to this gene, BCL2 is another mutated gene that inhibits programmed cell death. As such, cancerous cells proceed with their continued proliferation and leukemic cells are released from the tissue where they were generated.84 Hitzler et al reported that a mutation of the GATA1 gene in acute megakaryoblastic leukemia affects hematopoietic transcriptional factor. On the other hand, chromosomal translocation of t(7;11)(p15;p15) HSCs lead to the integration of genes, including HOXA9 and NUP98, which also leads to distortion in the transcriptional process of hematopoietic precursor cells.85 Aberration of the transcriptional process in these cells induces abnormal cell proliferation, which may lead to AML.85 Overproliferation of lymphoblasts within bone marrow can also result in the pathogenesis of ALL.8,49

Emphasis on the eradication of hematologic malignancies has shifted from cytotoxic chemotherapy to donors immune cells.86 HSCT is utilized by 20,000 people in the US every year.87 It is applicable in treating patients with rare diseases, such as AML,22 ALL,8 CML, Burkitts lymphoma, HL, and NHL,11 and other hematologic malignancies.88 Although it serves as an alternative treatment method, HSCT still has a relapse risk among 40%80% of recipients.89 Both auto-HSCT and allo-HSCTare the main alternative cellular therapeutic methods to treat leukemia. Auto-HSCT is the appropriate and applicable therapeutic option for multiple myeloma1,18 and HL.11 Charles et al explained that auto-HSCT was more frequently utilized by European and North American countries than allo-HSCT to treat myeloma. A lower mortality rate for myeloma is seen with auto-HSCT. Auto-HSCT is an established treatment approach if myeloma is at an acute stage, but for older patients it requires extra improvement.90 The occurrence of GVHD among myeloma patients who undergo allo-HSCT is 50% compared to 5%20% of occurrence of auto-SCTpatients91 (). As such, fewer GVHD effects have been seen in auto-SCT n treating multiple myeloma and HL.11 Furthermore, in HIV-related lymphoma, auto-HSCT is considered an applicable therapeutic option in both relapsed HL1 and relapsed NHL patients.18,92

Comparison of allogeneic and autologous stem-cell transplantation with hematologic disorders. Autologous stem-cell transplantation has been utilized as a treatment protocol to treat MM and HL, due to its initial response, low relapse sensitivity, and positive positron-emission tomography (+PET). Patients at higher risk or progress of AML are treated with allo-HSCT. Chronic phase 1 (CP1), TKI intolerance, and blast crisis enables allo-HSCT to be a standard treatment option for the treatment of CML. Allo-HSCT is also a treatment option for NHL patients presenting with complete remission 1 and 2 (CR1 and CR2) indications and also relapse after auto-HSCT. Although they have graft-vs-leukemic toxic effects, they are a significant alternative cell-based therapy to treat hematologic malignancies.

Abbreviations: ALL, acute lymphocytic leukemia; AML, acute myeloid leukemia; CML, chronic myeloid leukaemia; HL, Hodgkins lymphoma; MM, multiple myeloma; NHL, non-HL.

On the other hand, allo-HSCT is a curative treatment approach for severe AML93 It has been confirmed that hematologic toxicity is lower in these recipient patients. Allo-HSCT has also been used as a treatment option for acute lymphoid leukemia and multiple myeloma.1,23,94 Though alternative treatments remain undefined, it is a valuable treatment tool for hematologic malignancies. Reduced-intensity conditioning after allo-HSCT has been seen in Spain.95 The toxic effect of allo-HSCT is associated with graft-vs-leukemia reactions. Chronic myelogenous leukemia patients show lower relapse rate than other allogeneically transplanted leukemia patients.96 The therapeutic landscape of CML has shifted dramatically with developments tyrosine-kinase inhibitors (TKIs), which target the BCRABL1 hybrid oncoprotein and block the constitutive activity of tyrosine kinase. The course of CML is typically triphasic, with an early indolent chronic phase (CP), followed by an accelerated phase and a blast (crisis phase (BP).97,98 For selection of appropriate TKIs, of CML patients should be tested for BCRABL1 kinasedomain mutation (mutation profile), disease phase, and patient comorbidities. For example, if the patient has such mutations as Y253H, E255K/V, or F359C, physicians recommend dasatinib or bosutinib as TKI. On the other hand, if patients are in an advanced disease phase (BP) or CML-CP (with T315I mutation), third-generation ponatinib is preferred over imatinib.99103 However, allo-HSCT remains a therapeutic option for patients in CML-CP whose CML has progressed after at least two TKIs and after trialing ponatinib therapy (for T315I mutation) to reduce the CML burden, and for the effectiveness of the transplantation.99,100,102 An improvement in immunologic tolerance and lowered GVHD effect mean allo-HSCT is the only curative treatment option for CML-BP104 (). Similarly to CML, highly complicated and severe AML is effectively treated with allo-HSCT.22 Complications of AML may lead to higher mortality and morbidity rates, which may be due to chronic GVHD among patients >50 years old.105 Pediatric ALL patients presenting with indications of higher relapse risk are treated (10% of treatment) with allo-HSCT.106

ALL patients who develop high relapse risk are indications for treatment with allo-HSCT.107 Allo-HSCT is a standard treatment method for ALL patients who are at higher risk.108 The use of allo-HSCT has lower toxicity in young patients.86 Allo-HSCT has lower relapse risk than auto-HSCT in multiple myeloma.18 Graft-vs-tumor reactions in hematologic malignancies depend on the donors T cells and donor lymphocyte infusions. The decision to perform allo-HSCT depends mainly on reduced intensity conditioning.109 Researchers haverecommended that the use of allo-HSCT should depend on strong clinical data; however, 28%49% of allo-HSCT patients develop relapse risks for disease.110 Moreover, allo-HSCT has been widely applied as a therapeutic option in both HL and NHL.11

SCs play a major role in cell-based therapy to treat both hematologic and nonhematologic malignant disorders. They are mainly involved in the application of transplantation. Adult SCs (bone-marrow SCs), PBSCs, and UCB are the major potential sources of HSCs used during SC transplantation. Similarly, apart from ethical issues associated with disruption of inner cell mass, ESCs and ELSCs are also sources of HSCs as a therapeutic option to be utilized in SC transplantation. The generation of HSCs from iPSCs through hematopoieticendothelial transition will be therapeutic options during times of inadequate availability of compatible donors. On the other hand, non-HSCs and MSCs are possible to use as coinfusion to support engraftment of transplants, hematologic reconstitution, and manage GVHD posttransplantation. Auto-HSCT and allo-HSCT are the major cellular therapeutic options to treat leukemia. The lower relapse risk, blast crisis, TKI-intolerant patients in the CP and at higher risk of disease, and higher relapse risk are indications to utilize allo-HSCT rather than auto-HSCT to treat different types of leukemia. Likewise, primary refractory sensitivity to relapse and positive PET are basic indications to prefer auto-HSCT to allo-HSCT in treating both multiple myeloma and HL. Therefore, allo-HSCT is a more applicable standard cellular therapeutic option than auto-HSCT for many classes of leukemia.

The authors acknowledge Mrs Yonas Akalu for proofreading, language editing, and grammatical corrections to improve this review article.

Allo-HSCT, allogeneic hematopoietic stemcell transplantation; auto-HSCT, autologous HSCT; CML, chronic myeloid leukemia; GVHD, graft-versus-host disease; ESCs, embryonic SCs; iPSCs, induced pluripotent SCs;MSCs, mesenchymal SCs; PBSCs, peripheral blood SCsVSELSCs, very small embryonic-like SCs.

All authors made a significant contribution to the work reported, whether in conception, study design, execution, acquisition of data, analysis, interpretation, or all those areas; took part in drafting, revising, or critically reviewing the article; gave final approval to the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.

The authors declare that they have no competing interests.

48. !!! INVALID CITATION !!!

Read the rest here:
Role of Stem-Cell Transplantation in Leukemia Treatment

Nano-Improvements to Rheumatoid Arthritis Stem Cell Therapy Show Success – AZoNano

An article published in the journal Biomaterials shows that [emailprotected]2 nanoparticles (NPs) synthesized with a short bacteriophage-selected mesenchymal stem cell(MSC) targeting peptide allowed the MSCs to take up these NPs. NP-modified MSCs produced greatly improved therapy of Rheumatoid Arthritis(RA) using stem cells.

Study:Highly effective rheumatoid arthritis therapy by peptide-promoted nanomodification of mesenchymal stem cells. Image Credit:Emily frost/Shutterstock.com

RA, which is marked by progressive joint degeneration andsynovial inflammation, is one ofthe primary widespread inflammatory arthritis thataffectsaround 1 % of the global population, however, it currently lacks an effective treatment.

Glucocorticoids (GCs), disease-modifying anti-rheumatic drugs (DMARDs) and non-steroidal anti-inflammatory drugs (NSAIDs)are the three maintypes of medicationscurrently used in clinical practice.

GCs and NSAIDscan help with joint pain and stiffness, but they may cause side effects such asheart problems, osteoporosis, infections andgastric ulcers.

Standard DMARDs, like methotrexate (MTX), can lessen swelling by inhibiting the synthesis of pro-inflammatory cytokines and have little effect on cartilage degeneration. MTX, on the other hand, has a short plasma half-lifeand a poor concentration of the drug in the inflammatory region of the body.

Other side effects may also include liver and kidney damage, bone marrow depletion, and gastrointestinal problems. Biological DMARDs have been rapidly developed in recent years, thoughtheir action slows the progression of structural damage by reducing inflammation and have issues including drug resistance and the potential to cause significant infections and malignant tumors.

Multilineage differentiation, inflammatory site and immunomodulationhoming are all features of MSCs. These distinctivecharacteristicsallowMSCs to become apotential treatmentfora variety ofinflammatory and degenerativediseases, including the treatment of RA,through cell therapy. Unfortunately, over 50 % of patients do not react to MSC treatment, and the therapeutic benefit of MSCs is only temporary.

Firstly, MSCs are susceptible to the inflammatory milieu and so lose their functions of immune-regulationwhen disclosed in an inflamed joint. Reactive oxygen species (ROS) are thought to be engaged in the inflammation development of RA and hence damaging to MSCs, as seen by the gradualdecline in the quantity of MSCs in RA patients' synovial fluid.

Secondly, while the direct impacts of MSCs on tissue regeneration in RA are unknown, an evidentclinical experiment found that MSC injections increased hyaline cartilage regeneration in RA patients. Nevertheless, the unregulated distinction of MSCs can alsoresult in the development of tumors andthe inability of cartilage repair.

As a result, it is important for an optimal stem cell strategy to include MSCs that have the ability to preserve their bio functions and chondrogenically develop to regenerate cartilage under the oxidative stress caused by RA.

According to thisstudy, RA therapy could be enhanced byshort targeting peptide-promoted nanomodification of MSCs. To begin with, [emailprotected]2 NPs wereproduced due to some of theirelements' appealing features. Mn and Cu both are critical trace components in the human body, and they play a keyrole in the production of natural Mnsuperoxide dismutase (SOD) and Cu-ZnSOD, respectively.

Cu and Mn can also encourage stem cell chondrogenesis. The study further explains the modification of [emailprotected]2 NPs with MSC-targeting peptides to increase the passage of the nanoparticles into MSCs since transporting nanomaterials into modifications of MSCs is still a difficult task.

To make [emailprotected]2/MET NPs, [emailprotected]2 NPs were injected with metformin. Lastly, MSCs were allowedto take up these NPs and utilizethem to effectively limit synovial inflammation and maintain cartilage structure, alleviating arthritic symptoms greatly.

This study demonstrates that VCMM-MCSs werecreated by engineering MSCs with catalase (CAT) and superoxide dismutase (SOD)- like activity using dynamically MSC-targeting [emailprotected]2/MET NPs.

The biological features of these cells required in stem cell treatment, such as chondrogenesis, anti-inflammation, cell migration, and increased survival under oxidative stress, were improved by VCMM-MCSs.

Consequently, the VCMM-MSCs injections reduced cartilage damage andsynovial hyperplasiain adjuvant-induced arthritis (AIA) as well as collagen-induced arthritis (CIA) models, substantially reducing arthritic symptoms. Since oxidative stress is present in numerous degenerative and inflammatory disorders, this strategy of altering MSCs with NPs could be applied to treat a number of other disorders as well as to achieve faster tissue healing using stem cell therapy.

Lu, Y., Li, Z. et al. (2022). Highly effective rheumatoid arthritis therapy by peptide-promoted nanomodification of mesenchymal stem cells. Biomaterials. Available at: https://www.sciencedirect.com/science/article/pii/S0142961222001132?via%3Dihub

Disclaimer: The views expressed here are those of the author expressed in their private capacity and do not necessarily represent the views of AZoM.com Limited T/A AZoNetwork the owner and operator of this website. This disclaimer forms part of the Terms and conditions of use of this website.

Visit link:
Nano-Improvements to Rheumatoid Arthritis Stem Cell Therapy Show Success - AZoNano

Lineage Announces Pipeline Expansion to Include Auditory Neuronal Cell Therapy for Treatment of Hearing Loss – Yahoo Finance

Expansion of Pipeline Into a Third Neuronal Cell Type Builds on Existing Capabilities

Intellectual Property Has Been Filed Covering Composition and Methods for Generating Auditory Neuronal Progenitors

Hearing Loss Afflicts More Than 5% of the Population; More Than 430 Million People

CARLSBAD, Calif., March 21, 2022--(BUSINESS WIRE)--Lineage Cell Therapeutics, Inc. (NYSE American and TASE: LCTX), a clinical-stage biotechnology company developing allogeneic cell therapies for unmet medical needs, today announced that the Company is expanding its novel cell therapy pipeline to include a new investigational product candidate, an auditory neuronal cell transplant for the treatment of hearing loss, with an initial focus on the treatment of auditory neuropathy spectrum disorders. To support this new therapeutic effort, Lineage has filed for intellectual property covering the composition and methods for generating auditory neuronal progenitors which may be capable of functioning as sensory neurons and the connecting neuronal ganglion cells of the ear, and to methods of treatment that employ these cells for the potential treatment of auditory neuropathy. According to the World Health Organization, hearing loss currently afflicts over 5% of the worlds population, or more than 430 million people, and by 2050 it is estimated that one in every ten people, or more than 700 million people, will have disabling hearing loss.

"Hearing loss is a major sensory deficit which affects an enormous number of individuals worldwide, yet current approaches leave much room for improvement. I am pleased to be advising Lineage and providing insights and experience in the launch of this new endeavor and working toward developing cell-based solutions for this condition," stated Stefan Heller, Ph.D., Edward C. and Amy H. Sewall Professor, Stanford University School of Medicine, Department of Otolaryngology Head & Neck Surgery and Institute for Stem Cell Biology and Regenerative Medicine ISCBRM.

Story continues

"We are excited to announce this new, internally-developed initiative for Lineage, and to do it so quickly following the partnership we announced with Roche and Genentech for our lead program, OpRegen, in a deal worth up to $670M USD," added Brian Culley, Lineage CEO. "Many patients with sensorineural hearing loss are poorly addressed, cannot benefit from cochlear implants, and/or have no FDA-approved treatment options. Similar to OpRegen, which has demonstrated to be able to replace and restore retinal pigment epithelium cells in patients with vision loss, and OPC1, which similarly replaces oligodendrocytes for the treatment of spinal cord injury, replacing auditory neurons or augmenting an existing but damaged auditory neuron population may provide a benefit beyond the reach of alternate approaches such as prostheses. We believe auditory neuronal transplants represent a unique opportunity to leverage our knowhow and capabilities in cellular differentiation into a fourth indication with a large unmet need. In addition to the speed with which the team created this new program from our internal technology, we have done so with a modest investment of capital so far, because we were able to take advantage of our established manufacturing infrastructure and broad knowhow in the expansion and differentiation of pluripotent cells. This is another example of the efficiency and versatility of our technology platform, which is gaining broader awareness, and which offers us a favorable competitive position in the emerging fields of regenerative medicine and anti-aging technologies."

Auditory neuropathy is a hearing disorder in which the inner ear successfully detects sound but has a problem with sending signals from the ear to the brain. Current state of the art medical knowledge suggests that auditory neuropathies play a substantial role in hearing impairments and deafness. Hearing depends on a series of complex steps that change sound waves in the air into electrical signals. The auditory nerve then carries these signals to the brain. Outer hair cells help amplify sound vibrations entering the inner ear from the middle ear. When hearing is working normally, the inner hair cells convert these vibrations into electrical signals that travel as nerve impulses to the brain, where the brain interprets the impulses as sound. Auditory neuropathy can be caused by a number of factors including: (i) damage to the auditory neurons that transmit sound information from the inner hair cells specialized sensory cells in the inner ear to the brain; (ii) damage to the inner hair cells themselves; (iii) inherited genes with mutations or suffering damage to the auditory system, either of which may result in faulty connections between the inner hair cells and the auditory nerve, which leads from the inner ear to the brain; or (iv) damage to the auditory nerve itself. Researchers are still seeking effective treatments for those affected with auditory neuropathy.

About Lineage Cell Therapeutics, Inc.

Lineage Cell Therapeutics is a clinical-stage biotechnology company developing novel cell therapies for unmet medical needs. Lineages programs are based on its robust proprietary cell-based therapy platform and associated in-house development and manufacturing capabilities. With this platform Lineage develops and manufactures specialized, terminally differentiated human cells from its pluripotent and progenitor cell starting materials. These differentiated cells are developed to either replace or support cells that are dysfunctional or absent due to degenerative disease or traumatic injury or administered as a means of helping the body mount an effective immune response to cancer. Lineages clinical programs are in markets with billion dollar opportunities and include four allogeneic ("off-the-shelf") product candidates: (i) OpRegen, a retinal pigment epithelium transplant therapy in Phase 1/2a development for the treatment of dry age-related macular degeneration, which is now being developed under a worldwide collaboration with Roche and Genentech, a member of the Roche Group; (ii) OPC1, an oligodendrocyte progenitor cell therapy in Phase 1/2a development for the treatment of acute spinal cord injuries; (iii) VAC2, a dendritic cell therapy produced from Lineages VAC technology platform for immuno-oncology and infectious disease, currently in Phase 1 clinical development for the treatment of non-small cell lung cancer and (iv) ANP1, an auditory neuronal progenitor cell therapy for the potential treatment of auditory neuropathy. For more information, please visit http://www.lineagecell.com or follow the Company on Twitter @LineageCell.

Forward-Looking Statements

Lineage cautions you that all statements, other than statements of historical facts, contained in this press release, are forward-looking statements. Forward-looking statements, in some cases, can be identified by terms such as "believe," "aim," "may," "will," "estimate," "continue," "anticipate," "design," "intend," "expect," "could," "can," "plan," "potential," "predict," "seek," "should," "would," "contemplate," "project," "target," "tend to," or the negative version of these words and similar expressions. Such statements include, but are not limited to, statements relating to the collaboration and license agreement with Roche and Genentech and activities expected to occur thereunder, the upfront, milestone and royalty consideration payable to Lineage and Lineages planned use of proceeds therefrom; the potential benefits of treatment with OpRegen, the potential success of other existing partnerships and collaborations, the broad potential for Lineages regenerative medicine platform and Lineages ability to expand the same; Lineages plans to advance its spinal cord injury, oncology and auditory neuron programs and announce new disease settings where it plans to deploy its technology; the projected timing of milestones of future studies, including their initiation and completion, the projected timing of interactions with the FDA to discuss product designation, manufacturing plans and improvements, and later-stage clinical development; the potential opportunities for the establishment or expansion of strategic partnerships and collaborations and the timing thereof, and the potential for Lineages investigational allogeneic cell therapies to generate clinical outcomes beyond the reach of traditional methods and provide safe and effective treatment for multiple, diverse serious or life threatening conditions. Forward-looking statements involve known and unknown risks, uncertainties and other factors that may cause Lineages actual results, performance or achievements to be materially different from future results, performance or achievements expressed or implied by the forward-looking statements in this press release, including, but not limited to, the risk that competing alternative therapies may adversely impact the commercial potential of OpRegen, which could materially adversely affect the milestone and royalty payments payable to Lineage under the collaboration and license agreement, the risk that Roche and Genentech may not be successful in completing further clinical trials for OpRegen and/or obtaining regulatory approval for OpRegen in any particular jurisdiction, the risk that Lineage might not succeed in developing products and technologies that are useful in medicine and demonstrate the requisite safety and efficacy to achieve regulatory approval in accordance with its projected timing, or at all; the risk that Lineage may not be able to manufacture sufficient clinical and, if approved, commercial quantities of its product candidates in accordance with current good manufacturing practice; the risks related to Lineages dependence on other third parties, and Lineages ability to establish and maintain its collaborations with these third parties; the risk that government-imposed bans or restrictions and religious, moral, and ethical concerns about the use of hES cells could prevent Lineage or its partners from developing and successfully marketing its stem cell product candidates; the risk that Lineages intellectual property may be insufficient to protect its products; the risk that the COVID-19 pandemic or geopolitical events may directly or indirectly cause significant delays in and substantially increase the cost of development of Lineages product candidates, as well as heighten other risks and uncertainties related to Lineages business and operations; risks and uncertainties inherent in Lineages business and other risks discussed in Lineages filings with the Securities and Exchange Commission (SEC). Lineages forward-looking statements are based upon its current expectations and involve assumptions that may never materialize or may prove to be incorrect. All forward-looking statements are expressly qualified in their entirety by these cautionary statements. Further information regarding these and other risks is included under the heading "Risk Factors" in Lineages periodic reports with the SEC, including Lineages most recent Annual Report on Form 10-K and Quarterly Report on Form 10-Q filed with the SEC and its other reports, which are available from the SECs website. You are cautioned not to place undue reliance on forward-looking statements, which speak only as of the date on which they were made. Lineage undertakes no obligation to update such statements to reflect events that occur or circumstances that exist after the date on which they were made, except as required by law.

View source version on businesswire.com: https://www.businesswire.com/news/home/20220321005294/en/

Contacts

Lineage Cell Therapeutics, Inc. IR Ioana C. Hone (ir@lineagecell.com) (442) 287-8963

Solebury Trout IR Mike Biega (Mbiega@soleburytrout.com) (617) 221-9660

Russo Partners Media Relations Nic Johnson or David Schull Nic.johnson@russopartnersllc.com David.schull@russopartnersllc.com (212) 845-4242

Read the original here:
Lineage Announces Pipeline Expansion to Include Auditory Neuronal Cell Therapy for Treatment of Hearing Loss - Yahoo Finance