Author Archives: admin


Mogrify Raises Additional $16M To Advance Its Mission To Transform The Development Of Life-Saving Cell Therapies – Clinical Leader

Mogrify Ltd (Mogrify), a UK company aiming to transform the development of life-saving cell therapies, recently announced the initial close of its Series A funding. The Company raised $16M USD in this round, bringing the total investment to over $20M USD to date. The funding will support internal cell therapy programs, and the development and out-license of novel IP relating to cell conversions of broad therapeutic interest. Mogrify is also actively recruiting, and will increase headcount to 60 scientific, operational and commercial staff located at its state-of-the-art facility on Cambridge Science Park.

The funding round was led by existing investor Ahren Innovation Capital (Ahren), an investment fund co-founded by leading UK scientific entrepreneurs, supporting transformational companies at the cutting edge of deep science and deep tech. Parkwalk, the largest EIS growth fund manager, backing businesses with IP-protected innovations creating solutions to real-world challenges, 24Haymarket, an early investor in Mogrify and a prolific early-stage investment syndicate in deep technology and the life sciences, and the University of Bristol Enterprise Fund III, also contributed to the fundraise.

Mogrify has developed a proprietary direct cellular conversion technology, which makes it possible to transform (transmogrify) any mature human cell type into any other without going through a pluripotent stem cell- or progenitor cell-state. The Company is deploying this platform to develop novel cell therapies addressing musculoskeletal, auto-immune, cancer immunotherapy, ocular and respiratory diseases as well as generating a broad IP position relating to cell conversions that exhibit safety, efficacy and scalable manufacturing profiles suitable for development as cell therapies.

Mogrify is commercializing its technology platform via a model that includes development and out-license of internally developed cell therapy assets, development and license of novel cell conversion IP, and the formation of joint-ventures to exploit the platform and/or novel cell conversion IP in non-core areas.

Mogrify launched in February 2019, announcing $3.7M USD seed funding from Ahren, 24Haymarket and Dr. Darrin M. Disley, OBE and went on to secure grants from Innovate UK and SBRI Healthcare.

Mogrify also strengthened its management and scientific teams and relocated a 20-strong workforce to its new headquarters at TusParks Cambridge Bio-Innovation Centre on Cambridge Science Park in May. It has now begun recruiting up to 40 additional commercial, operational and scientific roles to support its expanding pipeline of internal programs, as well as supporting numerous biotech and pharma collaborators in developing novel IP to underpin existing and new cell therapy programs.

Dr. Darrin M. Disley, OBE, CEO, Mogrify, said: Following the recent announcement of Dr. Jane Osbourn, OBE, as Chair of Mogrify, I am delighted we have been able to make an initial close of this fundraising round, with the backing of both existing and new investors. Due to the significant interest, we have been able to secure this growth-funding without engaging in a protracted and distracting fund-raising process. Having now raised over $20M, we can focus on delivery of our business strategy with the support of an aligned investor group. We will continue to engage with high-caliber investors with computational biology and cell therapy domain expertise as part of our on-going investor relations and capital markets strategy.

Alice Newcombe-Ellis, Founder and Managing Partner, Ahren Innovation Capital, said: Mogrifys technology is well positioned to disrupt the global cell therapy market. The Company has grown rapidly since February, appointing a world-class management team and delivering strongly against its business plans. We look forward to supporting Mogrify as it continues to go from strength to strength.

Alastair Kilgour, Chief Investment Officer, Parkwalk, said: We are delighted to be supporting the team at Mogrify, many of whom have been involved successfully with companies we have previously invested in, in this investment round. The science and technology base Mogrify are building is truly unique and disruptive. If successful, the positive effect on patient outcomes across a wide range of diseases will be staggering.

Alice Newcombe-Ellis, Founder and Managing Partner of Ahren, and Alastair Kilgour, Chief Investment Officer, Parkwalk, both join Mogrifys board of directors along with Dr. Karin Schmitt, the Companys Chief Business Officer.

For more information, visit https://mogrify.co.uk/investors/

About MogrifyMogrify has developed a proprietary direct cellular conversion technology, which makes it possible to transform (transmogrify) any mature human cell type into any other without going through a pluripotent stem cell- or progenitor cell-state.

The platform takes a systematic big-data approach to identify, from next-generation sequencing and gene-regulatory networks, the transcription factors (in vitro) or small molecules (in vivo), needed to convert a cell. By bypassing the stem cell-stage of cell transformation, Mogrify simultaneously addresses challenges associated with efficacy, safety and scalability.

Mogrify is deploying this platform to develop novel cell therapies addressing musculoskeletal, auto-immune, cancer immunotherapy, ocular and respiratory diseases as well as generating a broad IP position relating to cell conversions that exhibit safety, efficacy and scalable manufacturing profiles suitable for development as cell therapies.

Uniquely positioned to address a cell therapy market estimated to be $35B USD by 2023, Mogrify is commercializing its technology via IP licensing, product development, and drug development. Based in Cambridge, UK, the Company has raised over $20M USD funding from Ahren Innovation Capital, Parkwalk, 24Haymarket, Dr. Darrin M. Disley, OBE and the University of Bristol Enterprise Fund III. For more information, visit http://www.mogrify.co.uk

About Ahren Innovation CapitalAhren LP is an investment fund that supports transformational companies at the cutting edge of deep science and deep tech. The technologies of its Founding Partners are today valued more than $100B combined.

A group of highly diverse, creative and original thinkers leading their domains, Ahren believes in taking considered risk that will deliver superior rewards capturing a generational opportunity to provide smart capital to deep technology pioneers.

With a philosophy espousing the importance of relationships and trust, Ahren provides long-term capital and support to exceptional founders and teams, empowering them to achieve the unimaginable.

Ahren Innovation Capital was founded by Alice Newcombe-Ellis, together with Science Partners Sir Shankar Balasubramanian, Professor John Daugman, Professor Zoubin Ghahramani, Professor Steve Jackson, Professor Andy Parker, Sir Venki Ramakrishnan, Lord Martin Rees and Sir Gregory Winter. For more information, visit http://www.ahreninnovationcapital.com

About ParkwalkParkwalk is the largest growth EIS fund manager, backing world-changing technologies emerging from the UKs leading universities and research institutions. With 250M of assets under management, it has invested in over 100 companies across its flagship Parkwalk Opportunities EIS Fund as well as the award-winning enterprise and innovation funds Parkwalk manages for the Universities of Cambridge, Oxford and Bristol.

Parkwalk invests in businesses creating solutions to real-world challenges, with IP-protected innovations, across a range of sectors including life sciences, AI, quantum computing, advanced materials, genomics, cleantech, future of mobility, MedTech and big data.

For more information, visit http://parkwalkadvisors.com

About 24Haymarket24Haymarket is a premium deal-by-deal investment platform focused on high-growth businesses, investing up to 5M in any company. 24Haymarkets Investor Network includes several highly experienced private equity and venture capital investors, seasoned entrepreneurs and senior operators. We invest our own capital in direct alignment with entrepreneurs and typically seek Board representation to actively support their growth agenda. Since its inception in 2011, 24Haymarket has invested in more than 50 high-growth businesses. For more information, visit http://www.24haymarket.com

The University of Bristol Enterprise Fund III (Managed By Parkwalk)The University of Bristol Enterprise Fund is an early stage investment fund backing scientific and technological companies spun out of the University of Bristol or being supported by the Universitys SETsquared incubator. For more information, visit http://parkwalkadvisors.com/fund/university-of-bristol-enterprise-fund.

View original post here:
Mogrify Raises Additional $16M To Advance Its Mission To Transform The Development Of Life-Saving Cell Therapies - Clinical Leader

LogicBio Therapeutics to Present New Data on Next Generation Capsid Development Program and GeneRide Platform Program at the European Society of Gene…

CAMBRIDGE, Mass., Oct. 16, 2019 (GLOBE NEWSWIRE) -- LogicBio Therapeutics Inc. (Nasdaq:LOGC), a genome editing company focused on developing medicines to durably treat rare diseases in pediatric patients, today announced upcoming presentations at the European Society of Gene and Cell Therapy (ESGCT) 27th Annual Congress, held in Barcelona, Spain, October 22-25, 2019.

We are thrilled to be presenting positive data on our Next Generation Capsid Development Program on the anniversary of our collaboration with Childrens Medical Research Institute of Australia, a leader in gene therapy, childhood cancer, embryology and neurological diseases. The goal of the collaboration is to develop novel, synthetic adeno-associated virus (AAV) capsids which are highly tropic for human tissues and optimized for manufacturing. These data give us further confidence that we can improve the performance of current AAV vectors, expanding our pipeline and strengthening our GeneRide platform, said Fred Chereau, CEO of LogicBio. Further, we are pleased to present additional preclinical data further supporting the durability of expression, compared to canonical gene therapy, in one of our GeneRide platform programs and to have been invited to speak on AAV manufacturing.

Panel PresentationTitle: AAV manufacturing: critical parameters influencing vector quality attributesPresenter: Matthias Hebben, Ph.D., VP, Technology Development, LogicBio Therapeutics (INV36)Session: 1d ATMP manufacturingSession date/time: October 23, 2019, 8:30-10:30 a.m. CEST

Poster PresentationsTitle: AAV development program: towards next generation of livertropic AAV variants (P025)Session date/time: October 23rd, 2019, 1:00-3:00 p.m. CEST

Title: Durability of factor IX expression in mice treated neonatally with a nuclease-free, promoterless, AAV-based gene therapy, GeneRide (P423)Session date/time: October 23rd, 2019, 1:00-3:00 p.m. CEST

Additional information on the meeting can be found on the ESGCT website: https://www.esgct.eu/home.aspx

About LogicBio TherapeuticsLogicBio Therapeutics is a genome editing company focused on developing medicines to durably treat rare diseases in pediatric patients with significant unmet medical needs using GeneRide, its proprietary technology platform. GeneRide enables the site-specific integration of a therapeutic transgene in a nuclease-free and promoterless approach by relying on the native process of homologous recombination to drive potential lifelong expression. Headquartered in Cambridge, Mass., LogicBio is committed to developing medicines that will transform the lives of pediatric patients and their families.

For more information, please visit http://www.logicbio.com.

Forward-Looking Statements

This press release contains forward-looking statements within the meaning of the federal securities laws. These are not statements of historical facts and are based on managements beliefs and assumptions and on information currently available. They are subject to risks and uncertainties that could cause the actual results and the implementation of the Companys plans to vary materially, including the risks associated with the initiation, cost, timing, progress and results of the Companys current and future research and development activities and preclinical studies and potential future clinical trials. These risks are discussed in the Companys filings with the U.S. Securities and Exchange Commission (SEC), including, without limitation, the Companys Annual Report on Form 10-K filed on April 1, 2019 with the SEC, and the Companys subsequent Quarterly Reports on Form 10-Q and other filings with the SEC. Except as required by law, the Company assumes no obligation to update these forward-looking statements publicly, even if new information becomes available in the future.

Contacts

Brian LuqueAssociate Director, Investor Relationsbluque@logicbio.com951-206-1200

Stephanie SimonTen Bridge Communicationsstephanie@tenbridgecommunications.com617-581-9333

Visit link:
LogicBio Therapeutics to Present New Data on Next Generation Capsid Development Program and GeneRide Platform Program at the European Society of Gene...

Dunbar CAR T-Cell Program brings advanced immunotherapy to cancer patients – WHAS11.com

LOUISVILLE, Ky. Cancer patients in Louisville and throughout the region soon will have access to some of the most advanced immunotherapy treatments available.

Louisville resident Thomas E. Dunbar has pledged $1 million to the University of Louisville to create a specialized center to provide chimeric antigen receptor positive T (CAR T) cell therapies to patients at the U of L James Graham Brown Cancer Center and other centers in the Midwest.

The new program will be named the Dunbar CAR T-Cell Program.

This gift will allow both kids and adults to be treated right here in Kentucky with the most innovative cell-based immunotherapy being developed, said Jason Chesney, M.D., Ph.D., director of the U of L Brown Cancer Center.

In CAR T-cell therapies, immune cells are extracted from the patients own blood and then are genetically modified to fight cancer. The modified cells are infused back into the patient where they fight the cancer and create long-term immunity to its recurrence.

In addition to dramatic treatment results, CAR T-cell immunotherapy leads to fewer toxic side effects than traditional chemotherapy.

Patients who have been treated with all the conventional therapies who then underwent treatment in clinical trials with CAR T cells had dramatic response rates. Eighty-three percent of kids in the original trial who had lethal, terminal B-cell acute lymphoblastic leukemia responded to this therapy, Chesney said.

The Dunbar CAR T-Cell Program will include laboratories for manufacturing the CAR T cells and will administer both FDA-approved and clinical-trial therapies to adult and pediatric cancer patients.

The goal is for the facilities to be fully functional and receiving patients by Sept. 30, 2020.

Tom Dunbars son, Evan, lost his battle to cancer with neuroblastoma in 2001 at the age of 6. In 2009, Wally Dunbar, Tom Dunbars father, lost his battle with melanoma.

Donor Tom Dunbar with his son, Evan

U of L Brown Cancer Center

This year, Toms physician wife, Stephanie Altobellis, M.D., helped identify his own cancer.

Kentucky is at ground zero, with the nations highest rates of cancer diagnosis and death, Tom Dunbar said. Its completely unacceptable. We have to lead the charge right here where the need is the greatest and we can do the most good. We need treatments that are not toxic. Watching our loved ones miserable with pain, often just from the treatments, and yet still die in front of us simply cant be the best that we can do.

To learn more about how CAR T-cell treatment works visit: uoflbrowncancercenter.org

Make it easy to keep up-to-date with more stories like this. Download the WHAS11 News app now. For Apple or Android users.

Have a news tip? Email assign@whas11.com, visit our Facebook page or Twitter feed.

Go here to see the original:
Dunbar CAR T-Cell Program brings advanced immunotherapy to cancer patients - WHAS11.com

GSK partners with Lyell on cell therapies development – BioPharma-Reporter.com

GlaxoSmithKline announced a five-year collaboration agreement with Lyell Immunopharma, a San Francisco, US-based company, working on methods to prevent inhibition of T cells by tumors and relapses due to loss of T cell functionality.

The agreement will see the two companies working on the advancement of GSKs cell therapy pipeline, specifically on GSK3377794, a potential treatment for multiple cancer types currently in Phase II clinical development, which targets the NY-ESO-1 antigen.

According to GSK, although the first two chimeric antigen receptor (CAR)-T cell therapies, Yescarta (axicabtagene ciloleucel) and Kymriah (tisagenlecleucel), have now reached the market, engineered T cells have not yet delivered strong clinical activity in common solid tumours.

Improving the fitness of T cells and delaying the onset of T cell exhaustion could help engineered T cell therapies become more effective, the company stated.

Further than GSKs cell therapy candidate, the research partnership will look to advance Lyells approach of enhancing initial T cell response against solid tumours into a platform technology for future cell and gene therapies development projects to treat rare types of cancer.

Lyells technology, according to Rick Klausner, the companys CEO, looks to tackle three barriers to T cell efficacy in solid tumours.

We are redefining the ways we prepare patient cells to be made into therapies, modulating cells functionality so that they maintain activity in the tumour microenvironment, and establishing methods of control to achieve specificity and safety for solid tumour-directed cell therapies, Klausner explained.

See more here:
GSK partners with Lyell on cell therapies development - BioPharma-Reporter.com

Ziopharm: 13 Failed Programs With 3 More On Deck – Seeking Alpha

In our most recent article, we focused on Iovance (IOVA), a cell therapy company that in our opinion has consistently generated positive data, has developed a commercializable product and is deploying a de-risked clinical strategy for a clear unmet need. As a result, we saw nearly 100% upside in Iovance's stock.

In this article, we highlight Ziopharm (ZIOP), a cell therapy company that we think represents the polar opposite to Iovance. Ziopharm's history is long and littered with suspended and abandoned programs. The company has not shown an ability to develop a commercial product. Looking at the company today, we believe its three main programs will be proven worthless in time. While we think Iovance can double from current levels, it is our view that Ziopharm represents a compelling short opportunity with 100% downside. Our price target is $0.

Below, we summarize our short thesis for Ziopharm:

Ziopharm's history is littered with programs that have failed clinical trials or failed to develop meaningfully

For context, Ziopharm was founded in 1998 and became public by entering into a reverse merger with an OTC-listed company in 2005. In September 2006, Ziopharm shares were up-lifted onto the NASDAQ. We think this should serve as a red flag. Companies that enter the market through reverse mergers often carry greater risk of fraud and tend to be lower quality. In fact, the SEC has issued a Bulletin cautioning investors against reverse mergers and later tightened the rules for reverse merger listings. While we have not uncovered any evidence of fraud, in our opinion Ziopharm is a classic example of a lower quality company.

It is also important to point out that since its founding, Ziopharm has not successfully brought a drug to market. When Ziopharm first came to the market, it aimed to advance its lead assets ZIO-101 and ZIO-201. ZIO-101 was an organic arsenic in development to treat hematologic cancer, and ZIO-201 was a formulation of isophosphoramide mustard in development to treat solid tumors. At the time of its reverse merger, Ziopharm claimed that peak sales for the two products "could approach $800 million."

Those peak sales numbers failed to materialize, and the company has spent the last 10 years developing these products with little to show in the end.

While these two products were the first programs to disappoint during Ziopharm's history, they were certainly not the last. A summary of the company's failed attempts at drug development is contained in the following chart.

Ziopharm has consistently burned cash and diluted shareholders

As none of its programs have generated meaningful cash through product sales, Ziopharm has repeatedly issued equity and diluted shareholders. In fact, since the company's up-listing to the NASDAQ in Sept. 2006, Ziopharm has burned nearly $500 mn of cash from operations (source: Bloomberg). Meanwhile, shares outstanding have increased by nearly ~12x (source: SEC filings). Not surprisingly, share performance has been weak. Since the up-listing, Ziopharm shares have lost 16% of their value as compared to the +417% return for the NASDAQ Biotech Index (NBI).

Our research indicates that Ziopharm's largest shareholder has ties to Brian Kaspar, the disgraced scientist who allegedly fabricated Avexis animal data and that the largest shareholder has a history of making statements about the company that we view as promotional

With Ziopharm's history of repeated failures and shareholder value destruction, it's not surprising that Ziopharm lacks significant institutional ownership from established healthcare funds. Nevertheless, we were surprised by its largest shareholder who is a vocal backer of the company. Ziopharm's largest shareholder is White Rock Capital, which is managed by Thomas ("Tom") Barton.

Tom Barton was an early-stage investor of Avexis and was involved when the company hired Brian Kaspar. Kaspar has been implicated, along with his brother, as one of the two scientists responsible for the data manipulation of Zolgensma, the Avexis gene therapy drug that Novartis (NYSE:NVS) acquired. The FDA has indicated that it is considering pursuing criminal penalties.

Tom Barton also has a history of making highly promotional statements about Ziopharm. For example, in June 2015, Barton told a reporter at the Boston Business Journal that "he [Barton] believes Ziopharm will be ripe for an acquisition for as much as $10 billion in the next year-and-a-half." At the time, Ziopharm had a market cap of $1.2 bn. A takeout at $10 bn would have represented an unprecedented 733% premium at the time of the article's publication. Instead, shareholders were "rewarded" with a 30% drop in share price and underperformance relative to the NASDAQ Biotech Index (NBI) over the next year and a half.

Source: Bloomberg

Tom Barton has mentioned Ziopharm in other interviews and was the source of an article from 2014 when he suggested that the article's author should take a look at Intrexon (XON) and "Intrexon cubs" such as Ziopharm, Synthetic Biologics (SYN) and Fibrocell Science Inc. (FCSC). Synthetic Biologics and Fibrocell Science both now have market capitalizations of under $30 mn.

In summary, our research indicates that Ziopharm came to the market with an inauspicious beginning, has a history of product development failures and shareholder dilution, and is backed by an investor whom we believe has a checkered history.

Ziopharm investors may not be aware of the shortcomings and potential issues regarding TCR T that make it different from successful CART T therapies

Today, Ziopharm bulls appear to be most excited about their TCR T therapy program. As we detailed in our Iovance article, the success of CAR T therapy in blood cancers was not repeated in solid tumors. Along with TIL therapy, TCR T has emerged as a potential cell-based technology to treat solid tumors. We are strong believers in Iovance's streamlined TIL approach and our research indicates it will likely be the next major revolution in cell-based cancer therapy for solid tumors. While TCR T could emerge as a viable alternative someday, we think Ziopharm is unlikely to be a major player in this market due to its risky approach. Some of these risks are innate to TCR Ts, but we think the major ones are tied to Ziopharm's methods.

For background, TCR T and CAR T therapy follow a very similar approach. In both, patient T-cells are extracted and combined with a DNA sequence that encodes for a protein that is designed to bind with a protein expressed by cancer cells. These cells are then usually expanded (allowed to multiply in vitro) and re-infused back into the patient, where they continue to multiply, detect, and eliminate the cancer.

The primary difference between the CAR T and TCR T approach lies in whether the engineered T cell product recognizes cancer proteins expressed on the surface of the cell (CAR T) or inside the cells (TCR T). Proteins called major histocompatibility complex (NYSEARCA:MHC) class I molecules present internalized proteins to the cellular surface for recognition by T-cells. The TCR T cells then bind with the MHC class I receptor along with the cancer protein ligand that is presented. If the T-cell recognizes the ligand as a foreign cancer protein (i.e., non-self), this is supposed to initiate an immediate response from the immune system against the non-self-antigen-presenting cell. This results in the cell's destruction (hopefully the cancer cell) and a further escalation of the immune response.

While TCR T may be a promising therapy for solid tumors and is similar to CAR T, it's important to note the TCR T approach remains to be validated. Importantly, there are a number of major obstacles that exist today.

1) "On-target, off tumor" toxicity could be substantial:

TCR T therapy appears to have higher sensitivity to its directed antigen compared to a CAR. While this is good in terms of potential potency, we believe it comes with increased safety risks. If the TCR T target is expressed on healthy cells, the targeting of healthy cells could result in substantial tissue destruction and toxicity. One of the frontrunners for TCR T therapy in the clinic, Adaptimmune (ADAP), has already seen five treatment-related deaths across four early-stage clinical trials; Additional literature has pointed out the "extreme care" that needs to be taken to ensure cross-reactivity with similar peptides does not occur following the death of a number of animal models.

Ziopharm is attempting to mitigate this risk by targeting its therapy on neoantigens, but there are no guarantees that the neoantigens will not also be expressed by some healthy cells. In addition, as we will go through later, we believe the neoantigen approach carries a risk of being less efficacious, is likely impractical from a manufacturing and cost perspective, and faces regulatory uncertainties.

2) Human leukocyte antigen (HLA) haplotype matching may drastically shrink the commercial opportunity:

The potency of TCR T relies on an interaction between the MHC and the peptides bound to it. In order to work, an "off-the-shelf" engineered T-cell product must be matched to the HLA alleles (genetic variations of a particular gene) of the patients it is treating to induce efficacy. There are three main HLA classes associated with MHC class I proteins (HLA-A, HLA-B, and HLA-C) and six main HLA classes associated with MHC class II proteins (HLA-DPA1, HLA-DPB1, HLA-DQA1, HLA-DQB1, HLA-DRA, and HLA-DRB1). Additionally, each HLA class has different mutational combinations that are donated by each parent (for example, HLA-A can be further subdivided into HLA-A*0201, HLA-A*0301, HLA-A*2402, among many other combinations of mutational variations of the HLA-A gene).

This is very problematic for TCR T therapy as therapeutic TCR can only be used in patients who express the appropriate HLA alleles. In addition to limiting the population from an efficacy perspective, if HLA alleles are mispaired, we believe patients risk developing potentially fatal graft-versus-host disease (GVHD), which has already been seen in murine models. Based on discussion with industry experts, we believe the FDA is taking a cautious approach initially to TCR T regulation and treating each HLA-matched therapy for each cancer type as a separate drug. This means a full development program would be needed for each HLA subtype in each type of cancer (including full clinical trials).

The end result is that the addressable market for each TCR T therapy is actually quite small given the number of TAM cuts that are needed for each therapy. For example, the most common HLA allele in white populations, HLA-A*0201, still only accounts for roughly 45% of the total white population (importantly, the US is only ~60% white and is becoming more heterogeneous over time).

3) TCRs have difficulty eradicating metastatic tumors because of the immunosuppressive tumor microenvironment:

Tumor cells are known to inhibit the expression of T-cell trafficking signals. Additionally, competition for glucose within the tumor frequently induces a hypoxemic state, thereby leading to suppressed T-cell activation.

We believe Ziopharm's "solutions" to address the above TCR T problems are likely to come with efficacy, efficacy, manufacturing time, and possible safety trade-offs

To date, Ziopharm has disclosed two main strategies to advance TCR T. The first is a partnership with the NCI to use Ziopharm's non-viral Sleeping Beauty cell therapy platform in creating personalized, neoantigen-directed TCRs. The second is a neoantigen "hotspot" (KRAS, p52, EGFR) program that appears to be moving forward internally within Ziopharm. Our research into these programs leads us to believe that the strategies are likely to add incremental risk to an already unproven and highly risky approach. As a result, we think Ziopharm's TCR T program is unlikely to work and, therefore, is worthless. Below we describe the basis for our views on these two strategies.

1) A partnership with the NCI for neoantigen-directed TCR T:

An investigator-sponsored trial has been started that is investigating the use of TCRs directed against neoantigens in glioblastoma, non-small cell lung cancer, breast cancer, and gastrointestinal/GU cancer. A neoantigen is a protein that results from a random somatic mutation that occurs in individual tumors and varies both between patients and within patients from time to time. Since this protein is not an endogenous protein normally made under any other circumstance, the hope is that by targeting these neoantigens, the risk of "on-target, off-tumor" binding can be eliminated.

However, in our view, such an approach would risk trading off substantial efficacy, would be very expensive, and would result in commercially unfeasible manufacturing time with today's technology. In order to identify unique tumor antigens, solid human tumors would require sequencing of the whole genome of each individual tumor in order to identify mutated genes and choose a motif of proteins that are presented by the HLA alleles. Tumor heterogeneity can thus result in mixed results depending on how many mutations the therapy "captures" when produced, reducing the efficacy potential. Additionally, each therapy is technically unique to the patient, which is problematic as there is no framework within the FDA to evaluate such a drug. This substantially increases regulatory risk in our view. The end result is summarized in neoantigen TCR T review papers that describe a personalized neoantigen approach as representing " a massive undertaking for the care of a single patients, at a time when there is an increasing focus on value in cancer care."

Perhaps not surprisingly, Ziopharm has never discussed how such a therapy is even close to being commercially viable. Recall that TIL therapy had remained an interesting academic experiment (but not a commercial drug) for nearly 20 years largely because the "vein-to-vein" manufacturing process took up to eight weeks before Iovance's innovations reduced this process to approximately three weeks. Based on our conversation with Ziopharm, it is our understanding that the neoantigen screening phase will take 4-6 months (17-26 weeks) per patient enrolled. In other words, this screening process is 5-9 times longer than what is viewed as commercially viable as a "vein-to-vein" time with both TIL and CAR T therapy. We believe that after factoring in patients who have progressed during the screening process and are no longer eligible or alive to receive therapy, this approach will result in a very low ORR rate on an ITT-basis.

Per clinicaltrials.gov, the trial is expected to enroll 200 evaluable patients across the four tumor types (50 per tumor) in 2-4 years. Since this trial is open-label, we would not be surprised if Ziopharm pushes the NCI to look opportunistically to report early-stage data in order to try and convince investors of the platform's activity. However, unless Ziopharm pre-specifies when the trial readouts will occur, such an approach would confound the unblinded nature of the trial (this happens by continuously "looking" at the evolving data results until they happen to look interesting and then publishing on that) vs. a true drug effect signal. We believe the institutional biotech investing community will be unconvinced by such an outcome.

To sum it up, Ziopharm's neoantigen-directed approach that is currently in clinic through the NCI may be so specialized and patient-specific that it may not be economic for broad use and may also not be approvable by the FDA. Furthermore, when accounting for patients who progress during the very lengthy screening process, it may not be an effective treatment for aggressive cancer. The first phase 1 trial is expected to take 2-4 years to enroll, which we feel means commercialization is likely a decade away even if Ziopharm's approach is viable.

2) A neoantigen "hotspot" trial that we understand will be developed internally by Ziopharm:

Few details are known about how this trial will be designed, but we believe the theory is relatively straightforward. There are certain mutations that are associated with a number of different cancer types that produce a neoantigen, such as KRAS, p53, and EGFR. Ziopharm appears to be attempting to develop a TCR product directed against these neoantigens hotspots. We believe Ziopharm will face the same substantial challenges that we detailed above regarding neoantigens generally. Solid tumors are highly heterogeneous, so the expressed neoantigen may actually only be a percentage of the total tumor burden of the patient and thus TCR T may have limited efficacy in such a patient.

Furthermore, HLA alleles, as described earlier, would significantly reduce the total addressable market for each individual therapy if the company proceeds with developing these drugs using an "off-the-shelf" approach. A TCR T expert that we spoke with believes that the largest HLA haplotype (group of alleles inherited from a single parent) for KRAS (HLA-CW8) represented only ~10% of all U.S. patients with a KRAS mutation. Ziopharm bulls may not realize that this approach will likely result in a small market opportunity.

Another obstacle for Ziopharm arises because the development of a drug to target neoantigens such as KRAS has been notoriously difficult. Initially hyped expectations are now being tempered by more mature data with the risk that follow-up confirmation scans may further reduce ORR. Therefore, there is a high risk that Ziopharm will not be able to successfully design a product that reproducibly engages the KRAS binding pocket, or that the efficacy of targeting KRAS may disappoint relative to what we see as current lofty expectations.

Lastly, while the neoantigen approach to TCR T is designed to improve the safety profile of the product, the safety profile of this general approach is still an unknown, as is Ziopharm's specific product. In summary, we view this program as very unlikely to succeed and do not believe a program that has publicly disclosed so few details at such an early-stage of development should warrant any value being ascribed to it.

Ziopharm investors may not be aware of what we believe is Kite Pharma/Gilead's superior approach to TCR T

We believe Ziopharm will face stiff competition from competitor programs if the general TCR T approach is eventually validated. In particular, we would note that Kite Pharma/Gilead (GILD), the manufacturer that developed Yescarta, has an exclusive license with the NCI for the use of TCR-based product candidates directed against MAGE A3 and A3/A6 as well as an exclusive license from Steve Rosenberg's lab for TCR Ts targeting neoantigens. We believe many investors in Ziopharm are completely unaware of the viral-based neoantigen program that is currently in development at Gilead and that directly competes with Ziopharm's TCR T program. We believe Kite/Gilead's viral approach is well-established and could easily leverage the entire validated Kite/Gilead cell therapy development platform.

While Ziopharm loves to highlight its relationship with the Rosenberg lab, we believe the relationship between Rosenberg and Kite/Gilead must be much closer given Kite helped brandish Rosenberg's legacy by commercializing his CAR T approach, and since their formal partnership has lasted since 2012. This is in comparison to Ziopharm's relationship with TCR T that began in 2017 and is still in very early stages of development.

In terms of execution, we believe it's no contest on whether investors should bet on Ziopharm (who has not been able to get a product to market) vs. Kite/Gilead (who successfully developed and marketed Yescarta, along with many other blockbuster drugs) if they desire TCR T exposure.

Source: Zhang et al. 2019

In the end, a combination of very high clinical and execution risk, direct competition from validated cell therapy platforms, and manufacturing/commercial uncertainty leads us to ascribe $0 of value to the Ziopharm TCR T program.

Ziopharm investors may be placing too much emphasis on the company's hiring of Drew Deniger

Ziopharm bulls have pointed to the company's hiring of Drew Deniger as a significant positive, but we think bulls may have gotten over their skis. Before market open on July 3rd, 2019, Ziopharm announced the hiring of Dr. Drew Deniger to direct Ziopharm's TCR-T program. The announcement added $100 mn of market cap to Ziopharm's stock that day, and bulls have pointed to Deniger as a key hire and compared him as an "heir" to the Rosenberg lab and even claimed that he could win a Nobel prize:

While we recognize that Drew has research experience in cell therapy and worked with Steve Rosenberg, we believe bulls place far too much value on the hiring of him than they should. Deniger was still very early in his academic career when he was hired as head of Ziopharm's TCR T program. As far as we can tell, he has never held an academic professorship position of any kind. A search on Pubmed yields 15 total publications of which he's been an author, and 8 first author publications after spending 13 years in academic research (seven as a graduate student at MD Anderson and six as a post-doc fellow at the NCI). This is compared to a search of Steve Rosenberg which yields 654 publications.

Our research leads us to believe Ziopharm's non-viral approach may carry significant safety risks

While investors are most excited about Ziopharm's TCR T program, some seem to still cling to hopes that Ziopharm's earlier CAR T program will bear fruit. Despite an FDA clinical hold that has been ongoing for over a year, investors hope that the Clinical Hold for the 3rd generation, point-of-care CAR T program will be resolved and that the new Investigational New Drug (NYSE:IND) application in later stage cancers will save the program.

We think these hopes are misplaced. Instead, we anticipate Ziopharm's CAR T program is likely to fail to materialize into a meaningful, active program and will be discontinued.

First, we believe there are efficacy and safety issues inherent to Ziopharm's non-viral CAR T approach. Investors may not realize that Ziopharm's Sleeping Beauty "point-of-care" approach to CAR T has never been clinically validated.

In contrast, the CAR T viral vector approach as a treatment for blood cancers has been validated through the clinical success and approval of both Yescarta and Kymriah. It's important to understand the differences in the approaches. We believe a good starting point to understanding the risks to the Ziopharm CAR T program comes from knowing the six main manufacturing steps to the FDA-approved CAR T therapies, as seen in the chart below.

While approved CAR T therapies offer a highly efficacious option for patients with blood cancer, the logistical complexities of manufacturing and administering CAR T therapy (i.e., steps 2-4 in the above chart) have been a headwind to broad use. Manufacturing time (also known as "vein-to-vein" time) of Yescarta is ~17 days, and Kymriah is ~22 days. Many people believe that shorter vein-to-vein times would improve efficacy and commercial success as long as CAR activity is not sacrificed.

Ziopharm claims its non-viral approach can reduce the manufacturing time to less than two days and offer a "point-of-care" offering. This is also sometimes referred to as the "third-generation CAR T" program or "Rapid Personalized Manufacturing (RPM)" process by the company.

While this may seem impressive on the surface, the details of how Ziopharm reduces the traditional CAR T manufacturing method to a two-day process are highly concerning to us. It appears that Ziopharm shortcuts at least two of the six steps in the viral CAR T process that is currently used:

Additionally, we believe Ziopharm may also be attempting to eliminate the lymphodepletion portion of the CAR T process, which we think also poses risks by reducing the ability for the CAR T cells to propagate appropriately in vivo:

We believe this is a very risky approach to CAR T. As the cell transformation process is never 100% efficient, cell expansion allows for the enrichment of CAR+ cells and therapeutic dosing of T-cells. This also enables a rapid in vivo (in person) expansion that peaks at roughly 14 days post-infusion.

While Ziopharm may save up-front time by electing not to expand the T-cells ex vivo (outside the body), we think this will almost certainly be offset by a longer in vivo expansion period. This also may not allow the drug to reach the peak CAR T concentration levels that are achievable with the on-market CAR T therapies. Efficacy may be further hindered if the lymphodepletion step is removed from the process as lymphodepletion is through to support T-cell engraftment by depleting populations of suppressive T regulatory (Treg) cells and eliminating competition for IL-7, IL-15, and IL-21. While Ziopharm attempts to circumvent the lymphodepletion step by tethering IL-15 onto the T cell's membranes, we see this as an unvalidated approach that may not be as effective as suppression of Tregs via administration of lymphodepleting agents such as cyclophosphamide and fludarabine (cy/flu). Due to these issues, we do not believe the patient will achieve CAR T activity that is equivalent to Yescarta and Kymriah.

Ziopharm's 3rd generation CAR T program remains on FDA Clinical Hold, perhaps due to the issues we highlighted above

We view FDA's decision to place the CAR T program on clinical hold on June 18th, 2018 due to poor cell viability as validation of our views. While Ziopharm has referred to this as a hold relating to the "chemistry, manufacturing and controls" (CMC), we think it is likely related to patient safety. We believe a product below the 70% viability threshold would not only trigger ethical concerns about injecting a product that may not have enough live cells to result in meaningful activity, but also that cellular debris caused by the dead cells could pose as a risk for potentially life-threatening occlusion and thrombosis (blood clots) especially in cancer patients who already have an extremely elevated risk of thrombosis.

In fact, we believe FDA makes it crystal clear in a regulatory consideration slide deck that "unreasonable or significant risk of illness or injury" is the driver for IND holds in cell therapy. Based on our discussions with industry experts, we understand that these two on-market competitors are well above 70% cell viability. In fact, we know that Yescarta production is 97% on-specification. Therefore, even if Ziopharm is able to consistently reach the 70% cell viability threshold, it certainly does not ensure that it will have a product with competitive safety and efficacy relative to existing on-market options. In fact, we believe the company's decision to pursue this CAR T strategy will result in an inferior product if it is ever able to obtain approval.

Based on Ziopharm's 2Q19 conference call, Ziopharm now claims it "understands" how to produce T-cells to reach the 70% viability threshold. Note that the company states only that it believes it knows how to do it and not that it has consistently achieved this important milestone.

Furthermore, Ziopharm's recent actions to move the CAR-T program into what we view as sicker patients suggest problems remain. The company is now highlighting a new IND for donor-derived CAR T therapy in transplant failure patients, specifically in patients who have relapsed with CD19+ leukemias and lymphomas.

Ziopharm bulls claim this is a signal that the company has improved the manufacturing process to meet the FDA's threshold for releasing the clinical hold. We disagree. We think the company is attempting to move its CAR-T program into a sicker patient population where the FDA is more lenient with regards to safety risk and that the company has not addressed the potential underlying safety issues behind the clinical hold. Additionally, this patient population represents a significantly smaller market opportunity than the 3rd generation CAR T trial that is on clinical hold. Therefore, while we're aware some investors may view this IND clearance as a positive signal for the 3rd generation program coming off hold soon, we believe they are entirely independent events.

Finally, the new IND is for donor-derived CAR T therapy, which we believe is very risky and poses its own unique safety concerns. By pursuing an allogenic approach with patient-derived cells, we think Ziopharm will now be faced with risks of inducing graft vs. host disease (GVHD), CAR T rejection, and off-target cleavage with gene editing that could result in oncogenic mutations of the T-cells.

Ziopharm tried the IL-12 program in melanoma and breast but stopped due to what we believe were efficacy and safety concerns

Our final section addresses Ziopharm's IL-12 program. Ziopharm has been working on the IL-12 program since at least 2014. To date, the program consists of all phase 1/2 programs and the data has been underwhelming in our view. Still, some investors believe the IL-12 program holds much promise. Below, we will describe the history of the program and reasons we do not place any value on IL-12.

The Ziopharm IL-12 program is an adenoviral vector for IL-12 expression that is controlled via an oral activator. While originally designed to treat melanoma and breast cancer, these programs were discontinued in these indications following what we believe to be unacceptable efficacy/safety profile in phase 2 studies. When the company announced plans to discontinue melanoma and pause the breast cancer program at a banking conference in early 2017, the company cited that the primary driver of its decision was "introduction and approval of highly promising new single and combination agents."

In effect, the company admitted that it didn't believe the IL-12 program in melanoma and breast cancer was competitive enough to pursue development in light of emerging competition and the new standard of care. However, we believe a safety issue also played into the decision, since 6 of the 9 breast cancer patients developed dangerous cytokine release syndrome. We believe the combination of a lack of efficacy versus standard of care and safety issues "deep sixed" the melanoma and breast cancer programs.

We believe the IL-12 program's focus on recurrent glioblastoma multiforme is likely to fail

Ziopharm then pivoted toward the backup indication of recurrent glioblastoma multiforme (rGBM) as the lead IL-12 program. We believe the reasons why GBM was reserved as a backup indication is clear. First, Ziopharm saw GBM as by far the smallest market for IL-12, as seen in investor presentations from 2014.

Second, GBM is an exceptionally difficult-to-treat cancer that has resulted in a large number of phase 3 failures even when earlier data has been positive. In fact, a recent study found that between January 2005 and December 2016, of eight completed phase 3 trials, only a single trial reported a positive outcome and implied a high degree of risk when trusting early-stage open-label data in GBM.

As a base case, we believe investors' risk adjustment to any GBM trial should be exceptionally high. Moreover, Ziopharm's IL-12 program should be viewed as even riskier since it previously failed to show a favorable risk/reward in easier-to-treat cancer.

In fact, the difficulties in GBM have already been seen in the IL-12 program. Despite originally licensing the IL-12 technology in 2011 and starting preclinical development in June 2013, the monotherapy GBM program has only managed to advance into phase 1. In fact, the monotherapy program was in phase 3 at one point; however, a CMC issue forced the program to be placed on an indefinite hold. As a result, Ziopharm reverted back to testing the drug in phase 1. We think this is yet another example of how Ziopharm hyped up a potential opportunity only to miss on execution.

We do not believe the IL-12 combination program holds much promise and may be unsafe

Continue reading here:
Ziopharm: 13 Failed Programs With 3 More On Deck - Seeking Alpha

Abeona Therapeutics Announces Publication of Positive Long-Term Data from Phase 1/2a Clinical Trial Evaluating EB-101 Gene Therapy for Recessive…

Sustained wound healing and favorable safety profile observed at three years post-treatment

Durable wound healing in large, disabling, chronic wounds

EB-101 associated with long-term molecular expression of Type VII collagen protein

NEW YORK and CLEVELAND, Oct. 15, 2019 (GLOBE NEWSWIRE) -- Abeona Therapeutics Inc. (ABEO), a fully-integrated leader in gene and cell therapy, today announced positive long term efficacy and safety results from its Phase 1/2a clinical trial evaluating EB-101, a gene-corrected cell therapy for recessive dystrophic epidermolysis bullosa (RDEB). Treatment with EB-101 resulted in sustained wound healing with a favorable safety profile at three years post-treatment. These long-term follow-up data in seven RDEB participants with 42 treated wounds were published in the peer-reviewed journal JCI Insight. Full text of the publication can be accessed here: https://insight.jci.org/articles/view/130554

These results bolster our belief that EB-101 is a safe and effective gene-corrected cell therapy capable of providing durable, long-lasting healing for the most disabling wounds in patients with RDEB, said Joo Siffert, M.D., Chief Executive Officer of Abeona. These results are particularly significant, as EB-101 treatment led to wound healing even in the most challenging to treat large and painful chronic wounds. Given the average RDEB chronic wound size is over 118 cm2, it is essential that potential new treatments are capable of addressing these wounds to improve quality of life. We thank our collaborators at Stanford and the patients who volunteered to participate in this study and look forward to building upon this strong clinical foundation with the initiation of the pivotal Phase 3 VIITALTM Study evaluating EB-101 for the treatment of RDEB.

Key Study findings include:

Wounds selected for treatment were present for a mean of 11.2 years (range 3-20 years)

Three years after treatment with EB-101, a majority of RDEB patients had sustained wound healing, with 80% (16/20) of wounds achieving 50% healing, and 70% (14/20) achieving 75%

Two years after treatment, only 1 of 6 untreated (17%), prospectively selected control wounds, had 50% healing

50% or greater wound healing was associated with no pain (0/16) and no itch (0/16) at treated sites three years post-treatment, compared with presence of pain in 53% (20/38) and itch in 61% (23/38) of wound sites at baseline

EB-101 was associated with long-term molecular expression of type VII collagen protein, which plays an important role in anchoring the dermal and epidermal layers of the skin

No serious treatment-related adverse events were observed during the three-year observation period

No replication competent virus was present at any time point

Researchers from Stanford University School of Medicine conducted the Phase 1/2a single-center, open-label clinical trial to evaluate the long-term wound healing and safety of EB-101 in seven adult patients with severe generalized RDEB and to assess patient-reported outcomes following treatment. Chronic open wounds, defined as wounds present and unhealed for at least 12 weeks, with a total area of at least 100 cm2, were required for enrollment. In the trial, gene-corrected EB-101 skin grafts (35 cm2 each) were transplanted onto six wound sites in each of the seven adult participants (n= 42 sites total) and wounds selected for treatment had been present for a mean of 11.2 years (range:3-20 years). Participants were followed for two to five years after transplantation of EB-101 and received standard of care therapies including iron supplementation and esophageal dilations during the study.

Story continues

Abeona is currently continuing preparations for the pivotal Phase 3 VIITALTM Study evaluating EB-101 for the treatment of RDEB pending the anticipated receipt of Chemical, Manufacturing and Controls (CMC) clearance from the U.S. Food and Drug Administration expected in Q4 2019.

About EB-101EB-101 is an investigational, autologous, gene-corrected cell therapy poised to enter late-stage development for the treatment of recessive dystrophic epidermolysis bullosa (RDEB), a rare connective tissue disorder without an approved therapy. Treatment with EB-101 involves using gene transfer to deliver COL7A1 genes into a patients own skin cells (keratinocytes) and transplanting them back to the patient to enable normal type VII collagen expression and facilitate wound healing. In the U.S., Abeona holds Regenerative Medicine Advanced Therapy, Breakthrough Therapy, and Rare Pediatric designations for EB-101 and Orphan Drug designation in both the U.S. and EU.

About Recessive Dystrophic Epidermolysis BullosaRecessive dystrophic epidermolysis bullosa, or RDEB, is a rare connective tissue disorder characterized by severe skin wounds that cause pain and can lead to systemic complications impacting the length and quality of life. People with RDEB have a defect in the COL7A1 gene, leaving them unable to produce functioning type VII collagen which is necessary to anchor the dermal and epidermal layers of the skin. There is currently no approved treatment for RDEB.

About Abeona TherapeuticsAbeona Therapeutics Inc. is a clinical-stage biopharmaceutical company developing gene and cell therapies for serious diseases. The Companys clinical programs include EB-101, its autologous, gene-corrected cell therapy for recessive dystrophic epidermolysis bullosa, as well as ABO-102 and ABO-101, novel AAV9-based gene therapies for Sanfilippo syndrome types A and B (MPS IIIA and MPS IIIB), respectively. The Companys portfolio of AAV9-based gene therapies also features ABO-202 and ABO-201 for CLN1 disease and CLN3 disease, respectively. Its preclinical assets include ABO-401, which uses the novel AIM AAV vector platform to address all mutations of cystic fibrosis. Abeona has received numerous regulatory designations from the FDA and EMA for its pipeline candidates and is the only company with Regenerative Medicine Advanced Therapy designation for two candidates (EB-101 and ABO-102). For more information, visit http://www.abeonatherapeutics.com.

Forward Looking StatementThis press release contains certain statements that are forward-looking within the meaning of Section 27A of the Securities Act of 1933, as amended, and Section 21E of the Securities Exchange Act of 1934, as amended, and that involve risks and uncertainties. These statements include statements about the timing for CMC clearance for the VIITAL trial and the Companys beliefs relating thereto, the Companys ability to provide additional transport stability data points in response to the FDA clinical hold letter and the timing thereof, the Companys belief that completion of its CMC work and the durable safety and efficacy data will ultimately be critical to support a future Biologics License Application, the ability of its management team to lead the Company and deliver on key strategies, the market opportunities for the Companys products and product candidates, and the Companys goals and objectives. We have attempted to identify forward-looking statements by such terminology as may, will, anticipate, believe, estimate, expect, intend, and similar expressions (as well as other words or expressions referencing future events, conditions or circumstances), which constitute and are intended to identify forward-looking statements. Actual results may differ materially from those indicated by such forward-looking statements as a result of various important factors, numerous risks and uncertainties, including but not limited to continued interest in our rare disease portfolio, our ability to enroll patients in clinical trials, the impact of competition, the ability to secure licenses for any technology that may be necessary to commercialize our products, the ability to achieve or obtain necessary regulatory approvals, the risk of whether or when the FDA will lift the clinical hold respecting the Companys planned Phase 3 clinical trial for EB-101, the impact of changes in the financial markets and global economic conditions, risks associated with data analysis and reporting, and other risks as maybe detailed from time to time in the Companys Annual Reports on Form 10-K and quarterly reports on Form 10-Q and other reports filed by the Company with the Securities and Exchange Commission. The Company undertakes no obligation to revise the forward-looking statements or to update them to reflect events or circumstances occurring after the date of this presentation, whether as a result of new information, future developments or otherwise, except as required by the federal securities laws.

Investor Contact:Sofia WarnerSenior Director, Investor RelationsAbeona Therapeutics+1 (646) 813-4710swarner@abeonatherapeutics.com

Media Contact:Scott SantiamoDirector, Corporate CommunicationsAbeona Therapeutics+1 (718) 344-5843ssantiamo@abeonatherapeutics.com

Go here to see the original:
Abeona Therapeutics Announces Publication of Positive Long-Term Data from Phase 1/2a Clinical Trial Evaluating EB-101 Gene Therapy for Recessive...

Joint Pain Injections Market By New Business Developments, And Top Companies | Allergan Plc, Pfizer, Sanofi – Healthcare News

New York City, NY: Oct 16, 2019 Published via (Wired Release) MarketResearch. Biz has a big history of serving detail research reports that help to create business opportunities through competitive study. One of the report we provide is Joint Pain Injections market analyzing market latest trends, identifying your rivals, validating opportunities, examining threats to your company, and changing your go-to-market and positioning strategy respectively.

The report on the Global Joint Pain Injections Market 2019 has been portrayed by experienced and knowledgeable market analysts and researchers. It is a phenomenal compilation of vital studies that explore the competitive landscape, segmentation, geographical exploration, and revenue, production, and consumption growth of the Global Joint Pain Injections market. Players can use the accurate market facts and figures and statistical analysis provided in the report to understand the current and upcoming growth of the Joint Pain Injections market. The Global Joint Pain Injections market size was XX Mn US$ and it is expected to reach XX Mn US$ by the end of 2028, with a CAGR of XX% during 2019-2028. The report includes CAGR, market shares, sales, Joint Pain Injections gross margin, value, volume, and other crucial market figures that give an exact view of the growth of the Joint Pain Injections market 2019.

Advanced primary and secondary research techniques and tools have been used by our Joint Pain Injections market analysts to compile this report. We use research sources and tools that are highly reliable and trustworthy. The Joint Pain Injections market report provides effective guidelines and recommendations for leading players to secure a position of strength in the worldwide market. Joint Pain Injections Market Emerging players can also use this research study to plan business strategies and get informed about upcoming challenges in the market. We provide an extensive competitive analysis that includes detailed company profiling of key players, a study on the Joint Pain Injections nature and characteristics of the competitors landscape, and other important studies.

For Detail Insight Go With This Free Sample Report Enabled With Respective Tables and Figures:https://marketresearch.biz/report/joint-pain-injections-market/request-sample

Major Players of the Global Joint Pain Injections Market 2019

Chugai Pharmaceutical Co Ltd, Sanofi, Zimmer Biomet Holdings Inc, Flexion Therapeutics Inc, Seikagaku Corporation, Anika Therapeutics Inc, Bioventus LLC, Ferring B.V., Allergan Plc. and Pfizer Inc

Market Segmentation:

Segmentation by Injection type: Corticosteroid Injections, Hyaluronic Acid Injections, Others (include, Platelet-rich plasma (PRP), Placental tissue matrix (PTM), etc.). Segmentation by joint type: Knee & Ankle, Hip Joint, Shoulder & Elbow, Facet Joints of the Spine, Others (include, Ball and socket, etc.). Segmentation by end-user: Hospital Pharmacies, Retail Pharmacies, Online Pharmacies

Global Joint Pain Injections Market: Regions and Countries

The research study comprises key results and discoveries of our monitoring and analysis of the Joint Pain Injections market 2019. We have provided crucial information points, which include expansions, divestments, new product launches, Joint Pain Injections partnerships, mergers, acquisitions, and other strategic initiatives taken by Joint Pain Injections market players. The report also serves price trends for region-wise markets and analysis of crucial market events on a regional as well as worldwide scale. Our analysis will guide you to take vital decisions in the Joint Pain Injections market relating to procurement, inventory, pricing, and production. We assist you to give a tough competition to your rivals by providing latest, actionable, real-time, and quick market information.

The report will help you to get how and whether or not the worldwide Joint Pain Injections market 2019 has become customer-centric. It offers details insights into customer needs and preferences for players to expand their brand value, connect better with their clients, and improve their sales in the Joint Pain Injections market globally. As part of our customer insights, we focus on product positioning, customers perception of Joint Pain Injections market competition, customer segmentation, customer needs, consumer buying behavior, and target customers.

Have Any Query Or Specific Requirement? Feel Free To Ask Our Industry Experts: https://marketresearch.biz/report/joint-pain-injections-market/#inquiry

Our competitor profiling comprises validation of distribution channels and products and services offered by and Joint Pain Injections financial performance of companies operating in the market 2019. We also give Porters Five Forces, PESTLE, and SWOT analysis to identify the competitive threat and study other aspects of the Joint Pain Injections market. The report provides strategic recommendations, competitor benchmarking for performance measurement, and study of partnership, merger, and acquisition targets and Joint Pain Injections industry best practices. It also offered an analysis of profitability and cost across the Joint Pain Injections industry value chain.

Core Objective of Joint Pain Injections Market:

Every firm in the Joint Pain Injections market has objectives but this market research report focus on the crucial objectives, so you can analysis about competition, future market, new products, and informative data that can raise your sales volume exponentially.

Size of the Joint Pain Injections market and growth rate factors.

Important changes in the future Joint Pain Injections Market.

Top worldwide competitors of the Market.

Scope and product outlook of Joint Pain Injections Market 2019-2028.

Developing regions with potential growth in the future.

Tough challenges and risk faced in market.

Global Joint Pain Injections top manufacturers profile and sales statistics.

Joint Pain Injections Market Dynamics 2019-2028.

Browse Full Summary of Joint Pain Injections Market Enabled with Respective Tables and Figures at: https://marketresearch.biz/report/joint-pain-injections-market/

Customize report of Global Joint Pain Injections Market as per client requirement also available. Please connect with our sales team (inquiry@marketresearch.biz), who will ensure that you get a report that suits your needs.

Contact Us:

Mr. Benni Johnson

Prudour Pvt. Ltd.

420 Lexington Avenue,

Suite 300 New York City, NY 10170,

United States

Tel: + 1-347-826-1876

Website: https://marketresearch.biz/

Go here to read the rest:
Joint Pain Injections Market By New Business Developments, And Top Companies | Allergan Plc, Pfizer, Sanofi - Healthcare News

Stem Cell Therapy Industry 2019 Global Market Size, Trends, Revenue, Growth Prospects, Key Companies and Forecast by 2023 – Markets Gazette

Bone marrow transplant is the most widely used stem-cell therapy, but some therapies derived from umbilical cord blood are also in use. Research is underway to develop various sources for stem cells, and to apply stem-cell treatments for neurodegenerative diseases and conditions, diabetes, heart disease, and other conditions.

With the ability of scientists to isolate and culture embryonic stem cells, and with scientists growing ability to create stem cells using somatic cell nuclear transfer and techniques to create induced pluripotent stem cells, controversy has crept in, both related to abortion politics and to human cloning. Additionally, efforts to market treatments based on transplant of stored umbilical cord blood have been controversial.

Click to Access Sample Pages @ https://www.orianresearch.com/request-sample/609805

Scope of the Report:

This report studies the Stem Cell Therapy market status and outlook of Global and major regions, from angles of players, countries, product types and end industries; this report analyzes the top players in global market, and splits the Stem Cell Therapy market by product type and applications/end industries.

USA is a huge market, and the total sum of the industry is more than 24 Million US dollars in 2014. At the same time, this industry continuously increases, with the development of global economy.

According to the research, the most potential market in the main countries of stem cell therapy industry is China, determined by the rising level of medical care. Besides, South America, Middle East should also be focused by the investors. They are the potential consumers of stem cell therapy.

Complete report on Stem Cell Therapy Market report spread across 139 pages, profiling 18 companies and supported with tables and figures.

Inquire more or share questions if any before the purchase on this report @ https://www.orianresearch.com/enquiry-before-buying/609805

Development policies and plans are discussed as well as manufacturing processes and cost structures are also analyzed. This report also states import/export consumption, supply and demand Figures, cost, price, revenue and gross margins. The report focuses on global major leading Stem Cell Therapy Industry players providing information such as company profiles, product picture and specification, capacity, production, price, cost, revenue and contact information. Upstream raw materials and equipment and downstream demand analysis is also carried out. The Stem Cell Therapy industry development trends and marketing channels are

Analysis of Stem Cell Therapy Industry Key Manufacturers:

Order a Copy of Global Stem Cell Therapy Market Report @ https://www.orianresearch.com/checkout/609805

This report studies the top producers and consumers, focuses on product capacity, production, value, consumption, market share and growth opportunity in these key regions, covering:

Market Segment by Type, covers

Market Segment by Applications, can be divided into

Table of Contents

There are 15 Chapters to deeply display the global Banknote-Printing Machine market.

Chapter 1, to describe Banknote-Printing Machine Introduction, product scope, market overview, market opportunities, market risk, market driving force

Chapter 2, to analyze the top manufacturers of Banknote-Printing Machine, with sales, revenue, and price of Banknote-Printing Machine, in 2016 and 2017;

Chapter 3, to display the competitive situation among the top manufacturers, with sales, revenue and market share in 2016 and 2017;

Chapter 4, to show the global market by regions, with sales, revenue and market share of Banknote-Printing Machine, for each region, from 2013 to 2018;

Chapter 5, 6, 7, 8 and 9, to analyze the market by countries, by type, by application and by manufacturers, with sales, revenue and market share by key countries in these regions;

Chapter 10 and 11, to show the market by type and application, with sales market share and growth rate by type, application, from 2013 to 2018;

Chapter 12, Banknote-Printing Machine market forecast, by regions, type and application, with sales and revenue, from 2018 to 2023;

Chapter 13, 14 and 15, to describe Banknote-Printing Machine sales channel, distributors, traders, dealers, Research Findings and Conclusion, appendix and data source.

Customization Service of the Report:Orian Research provides customization of reports as per your need. This report can be personalized to meet your requirements. Get in touch with our sales team, who will guarantee you to get a report that suits your necessities.

About Us:

Orian Research is one of the most comprehensive collections of market intelligence reports on the World Wide Web. Our reports repository boasts of over 500000+ industry and country research reports from over 100 top publishers. We continuously update our repository so as to provide our clients easy access to the worlds most complete and current database of expert insights on global industries, companies, and products. We also specialize in custom research in situations where our syndicate research offerings do not meet the specific requirements of our esteemed clients.

See original here:
Stem Cell Therapy Industry 2019 Global Market Size, Trends, Revenue, Growth Prospects, Key Companies and Forecast by 2023 - Markets Gazette

Synthetic presentation of noncanonical Wnt5a motif promotes mechanosensing-dependent differentiation of stem cells and regeneration – Science Advances

INTRODUCTION

Human mesenchymal stem cells (hMSCs) have become increasingly popular as a cell source for repairing bone and other musculoskeletal tissues due to their availability, accessibility, and multipotency (1). The lineage commitment of hMSCs has been shown to be regulated by various signals, including mechanical stimuli, soluble factors, and cellextracellular matrix (ECM) interactions, in their natural niche microenvironment (2). Meanwhile, the delayed reunion of fractured bone remains a major clinical complication of surgery despite advances in operative techniques. The direct injection of stem cells into bone defects generally leads to limited stem cell grafting and differentiation due to the lack of necessary biochemical cues in the defect sites (3). Developing osteoinductive biomaterial scaffolds by incorporating developmentally relevant signaling cues to support and guide the differentiation of implanted stem cells in situ will enhance the clinical outcomes of stem cell therapies (4). For instance, scaffolds have been decorated with a wide range of bioactive motifs, including growth factors, bioactive peptides, and small molecules, to boost their osteoinductivity (5, 6). In recent years, biomimetic/bioactive peptides have become increasingly popular as inductive motifs for the biofunctionalization of biomaterials due to their major advantages, including their ease of immobilization onto various biomaterials via bioconjugation, better stability than proteins, and low cost (7). We previously demonstrated that the decoration of a hydrogel scaffold with an N-cadherin mimetic peptide promoted the chondrogenesis or the osteogenesis of hMSCs by emulating the enhanced intercellular interactions (8, 9). Therefore, there is an acute demand for previously unidentified osteoinductive ligands, which can potentially be identified by examining developmentally relevant proteinaceous cues (10).

Wnt signaling pathways have been reported to be essential to many developmental events, especially osteogenesis and bone formation (11, 12). The canonical Wnt signaling pathways generally involve the preservation and nuclear translocation of -catenin, which further triggers the activation of downstream genes to initiate osteogenic differentiation (13). On the other hand, the -cateninindependent noncanonical Wnt signaling cascade is activated by noncanonical Wnt ligands, such as Wnt5a (14). Wnt5a activation has been reported to contribute to the regeneration of multiple tissues, including the articular cartilage, the colonic crypt, and the liver, via paracrine or autocrine regulation (1517). During in vitro osteogenesis, embryonic stem cells express Wnt5a early on, rather than canonical Wnt3a, and cells expressing Wnt5a or treated with exogenous Wnt5a show a substantially enhanced osteogenic yield (18). In trabecular bone and bone marrow, Wnt5a is mainly expressed and secreted by osteoblastic niche cells, including precursor cells, osteoblasts, osteoclasts, and osteocytes (19). Wnt5a has been reported to preserve the proliferation and differentiation potential of stem cells in bone marrow and induce osteoblast maturation (20). Wnt5a signaling is a substantial constituent of bone morphogenetic protein 2mediated osteoblastogenesis (21). Collectively, these findings indicate that Wnt5a is an essential cue for the development of bones.

Wnt5a signaling is mediated through the Disheveled (Dvl) segment polarity proteindependent mechanism, which activates the small guanosine triphosphatase (GTPase) RhoA and its effector ROCKs (Rho-associated protein kinases) (22). RhoA-ROCK signaling plays a major regulatory role in the mechanotransduction signaling of cells by promoting focal adhesion formation (16), stress fiber assembly (23), and actomyosin contractility, and all these cellular events have been shown to enhance the osteogenesis of mesenchymal stem cells (MSCs). However, to the best of our knowledge, no prior studies have capitalized on the osteoinductive potential of noncanonical Wnt ligands to functionalize biomaterials or investigated the efficacy of such a developmental biologyinspired strategy to enhance the osteogenesis of MSCs and associated bone formation.

Recently, a Wnt5a mimetic hexapeptide, Foxy5 (formyl-Met-Asp-Gly-Cys-Glu-Leu), was shown to trigger cytosolic calcium signaling by activating -cateninindependent noncanonical Wnt signaling (24), and Foxy5 has recently been tested in a phase 1 clinical trial for treating cancer (www.clinicalTrials.gov; NCT02020291) (25). Here, we hypothesized that the functionalization of biomaterials with Foxy5 peptide can promote the mechanosensing and osteogenesis of hMSCs by activating noncanonical Wnt signaling (Fig. 1A). Our findings showed that this synthetic presentation of Wnt5a mimetic ligand activated noncanonical Wnt signaling, elevated the intracellular calcium, and promoted the mechanotransduction and osteogenesis of hMSCs, resulting in the enhanced bone regeneration in vivo. These findings emphasize the significance of the biofunctionalization of biomaterial scaffolds with developmentally guiding cues to enhance in situ tissue regeneration via grafted stem cells (26, 27).

(A) Porous, Foxy5 + RGD peptideconjugated MeHA hydrogels were developed by conjugating cysteine-containing functional peptides to MeHA molecules. (B) The porous scaffolds were used to copresent the adhesive ligand (RGD) and noncanonical Wnt5aactivating ligand (Foxy5) to synergistically induce the osteogenic lineage commitment of stem cells both in vitro and in vivo. (C) Rat MSC (rMSC)seeded hydrogels were used to fill calvarial defects for regeneration. (D) Micrographs of the 3D porous hydrogels with 200-m pores. The inset shows the microstructure of the MeHA porous hydrogel; scale bar, 50 m. (E) The Young modulus of the DTT-crosslinked MeHA hydrogels was verified using the Mach-1 mechanical tester. (F) The live/dead staining of the hMSCs seeded in the porous MeHA hydrogels showed the uniform distribution of the cells in the hydrogels. (G) Viable cell metabolic activity in the RGD, Foxy5 + RGD, and Scram + RGD hydrogels on day 7 of culture was characterized by alamarBlue assay. Data are shown as the means SD (n = 3).

To determine the effect of the Wnt5a mimetic peptide (Foxy5) on cellular behaviors, we grafted methacrylated hyaluronic acid (MeHA) with Foxy5 peptide (MDGECL, 1 mM) and arginylglycylaspartic acid (RGD) peptide (GCGYGRGDSPG, 1 mM) via Michael addition between the cysteine thiols of the peptide and the methacrylate groups of the MeHA (Fig. 1A and fig. S1). The peptide-functionalized MeHA (degree of methacryloyl substitution or methacrylation degree = 100 or 30%) (fig. S1) was then cross-linked to fabricate a three-dimensional (3D) porous hydrogel scaffold or a 2D hydrogel substrate for subsequent experiments (Foxy5 + RGD) (Fig. 1, B to D). Control hydrogels were fabricated with either RGD peptide alone (RGD) or the combination of scramble-sequenced Foxy5 peptide and RGD (GEMDCL, 1 mM, Scram + RGD). The RGD peptide was included in all groups to promote cell adhesion. The average Young moduli of the hydrogels from the RGD, Foxy5 + RGD, and Scram + RGD groups were determined to be 11.26, 10.82, and 10.96 kPa, respectively, indicating similar hydrogel stiffness in all groups (Fig. 1E). The storage moduli and loss moduli acquired from the frequency sweep analysis were not significantly different among the RGD, Foxy5 + RGD, and Scram + RGD groups (fig. S2A). Similarly, the surface roughness and the stiffness of 2D ultraviolet (UV)cross-linked hydrogels are not significantly different (fig. S2B). Our previous experience shows that the photocrosslinked MeHA hydrogels are typically stiffer than the dithiothreitol (DTT)cross-linked MeHA hydrogels given the same macromer content and methacrylation degree. This can be due to the semirigid nature of hyaluronic acid (HA) backbone, which may hinder the efficient crosslinking of HA-grafted methacryloyl groups by bifunctional crosslinkers (e.g., DTT). Therefore, by using the MeHA with the lower (30%) methacrylation degree to fabricate 2D hydrogels, the average Young moduli of the photocrosslinked hydrogels with 667 s of UV radiation were not significantly different from those of the DTTcross-linked MeHA hydrogels (100% methacrylation degree) (fig. S2, C and D). We believe that the results acquired from the 2D hydrogel substrates are representative and comparable with the data obtained from the 3D macroporous hydrogels. The 3D porous hydrogels used in this work have large pore sizes of around a few hundred micrometers, which are significantly larger than that of cells. The cells seeded in these 3D hydrogels are essentially still residing on top of the curved surfaces of the pores and are interacting with a more 2D-like rather than 3D microenvironment, and this is similar to the lining of osteoblasts on the surface of porous trabecular bone.

To evaluate the cytocompatibility of the peptide-functionalized hydrogels, we seeded hMSCs into the porous hydrogel constructs and allowed them to adhere for 4 hours, followed by further culture in basal growth media for another 7 days. Live/dead staining after 7 days of culture revealed that the majority of the seeded stem cells were viable and uniformly adhered to the RGD, Foxy5 + RGD, and Scram + RGD porous hydrogels (Fig. 1F). The alamarBlue assay showed that the seeded cells in all groups maintained consistent and robust metabolic activity in the porous hydrogel scaffolds for 7 days (Fig. 1G). These results indicate that the conjugated bioactive Foxy5 peptide was noncytotoxic, consistent with previous reports (24).

To further investigate the molecular events mediated by the noncanonical Wnt5a mimetic Foxy5 peptide, we used immunofluorescence staining to examine the expression levels of integrin V, integrin 1, phosphorylated focal adhesion kinase (p-FAK), and ROCK2 (fig. S3), which are essential elements for mediating mechanotransduction and have been reported to promote the osteogenic differentiation of stem cells. The staining intensities of integrin V and integrin 1 in the hMSCs cultured in the Foxy5 + RGD group appeared slightly higher but were not statistically different compared with that in the RGD and Scram + RGD groups (fig. S3, A and B). The staining intensity of p-FAK in the Foxy5 + RGD group was 92 and 33% higher than those in the RGD and Scram + RGD groups, respectively (fig. S3C). Consistent with the elevated expression of focal adhesion complex components, the Foxy5 + RGD group also showed ROCK2 staining intensity 135 and 34% higher than those in the RGD and Scram + RGD groups, respectively, after 7 days of osteogenic culture (fig. S3D). This enhanced expression of p-FAK and ROCK2 supports our speculation that the Foxy5 peptide presented by the biomaterial facilitates the mechanotransduction of the cells by activating noncanonical Wnt signaling to up-regulate the expression of focal adhesion complex molecules (p-FAK) and mechanotransduction signaling molecules (ROCK2).

We next explored the molecular signaling events by which the presentation of Foxy5 peptide via the hydrogel scaffold increased the mechanotransduction and consequent osteogenic lineage commitment of hMSCs (Fig. 2A). Noncanonical Wnt5a signaling has been shown to regulate the signaling of the Rho family of GTPases, such as RhoA, and studies have shown that RhoA is essential for actin cytoskeletal stability and associated actomyosin contractility via its downstream effectors, including ROCKs (22). Therefore, to examine the contribution of RhoA signaling and actomyosin contractility to the osteogenic effect of Foxy5 peptide presentation, we added Y-27632, an inhibitor of ROCKs, and blebbistatin, an inhibitor of nonmuscle myosin II (NMII), to the media in the Foxy5 + RGD group during the 7 days of osteogenic culture. The inhibition of ROCK with Y-27632 completely abolished the up-regulated osteogenic gene expression in the Foxy5 + RGD hydrogels. Blocking NMII activity also led to significantly down-regulated expression of the osteogenic genes alkaline phosphatase (ALP), type I collagen, and osteopontin (OPN) compared with the corresponding levels in the RGD group (fig. S4). Specifically, after 7 days of osteogenic culture, the expression levels of type I collagen, ALP, RUNX2, and OPN in the Y-27632Foxy5 + RGD group were down-regulated by 80.20, 97.96, 71.10, and 97.70%, respectively, compared with those in the Foxy5 + RGD group. The expression levels of type I collagen, ALP, RUNX2, and OPN in the blebbistatinFoxy5 + RGD group were down-regulated by 86.23, 98.54, 24.24, and 87.48%, respectively, compared with those in the Foxy5 + RGD group. These findings suggest that the pro-osteogenic effect of the Foxy5 peptide presentation can be attributed to the activation of RhoA signaling and associated actomyosin contractility.

(A) Schematic illustration of the seeding of hMSCs on the Foxy5/Scram + RGD peptidefunctionalized 2D hydrogel substrate. (B) Gene expression level of the RhoA signaling cascade (Wnt5a coreceptor Dvl2, RhoA, ROCK), downstream mechano-effector (NMII), and major focal adhesion adaptor protein (vinculin) in hMSCs in 3D porous hydrogels conjugated with RGD peptide alone (RGD), Foxy5 and RGD peptide (Foxy5 + RGD), or scrambled Foxy5 peptide and RGD peptide (Scram + RGD), respectively, after 7 days of osteogenic culture (n = 9). (C) Representative micrographs of fluorescence staining for F-actin (red), nuclei (blue), and RhoA (green) in hMSCs cultured on the 2D RGD, Foxy5 + RGD, and Scram + RGD hydrogels. Quantification showed a significantly higher RhoA staining intensity in the Foxy5 + RGD group than in the RGD and Scram + RGD groups (n = 20). a.u., arbitrary units. (D) Western blot bands and quantification of the expression level of mechano-responsive kinases ROCK2 and p-FAK (phosphorylated at the Ser722 sites) in each group (RGD, Foxy5 + RGD, Scram + RGD). (E) Representative merged fluorescence and bright-field micrographs of intracellular calcium in hMSCs cultured on RGD, Foxy5 + RGD, and Scram + RGD 2D hydrogels (stained with Fura-AM). (F) Quantification showed a significantly higher intracellular calcium level in the Foxy5 + RGD group than in the RGD and Scram + RGD groups. Scale bars, 50 m. Data are shown as the means SD (n = 9). Statistical significance: *P < 0.05, **P < 0.01, and ***P < 0.001.

We further investigated the mechanism underlying the enhanced RhoA signaling and actomyosin contractility via Foxy5 peptidemediated noncanonical Wnt signaling. Gene expression analyses revealed the up-regulated expression of Dvl2, RhoA, ROCK2, vinculin, and NMII in the presence of conjugated Foxy5 peptides after 7 days of osteogenic culture (Fig. 2B). Specifically, the Foxy5 + RGD group showed Dvl2, RhoA, ROCK2, vinculin, and NMII expression levels that were increased by 43, 27, 65, 72, and 24%, respectively, compared with those in the RGD group. Meanwhile, the expression of ROCK1, which was speculated to contribute to F-actin instability, was down-regulated by 25% in the Foxy5 + RGD group compared with that in the RGD group. Furthermore, the expression of the canonical Wnt signalingrelated genes (Wnt3a, Frizzled 3, LRP5, LRP6, and -catenin) was significantly up-regulated in the Foxy5 + RGD group compared with that in the control groups (fig. S5), and this is consistent with a previous report showing that the activated noncanonical Wnt signaling up-regulated the expression of canonical Wnt signaling factors during osteoblastogenesis (28). The expression of these mechanotransduction-related genes and canonical Wnt signalingrelated genes in the Scram + RGD group was not significantly different from that in the RGD group (Fig. 2B and fig. S5). We further quantified the expression of RhoA, ROCK2, and p-FAK based on immunofluorescence staining and Western blot analysis. The Foxy5 + RGD hydrogels showed RhoA staining intensity 92 and 134% higher than that in the RGD group and Scram + RGD groups, respectively, after 7 days of osteogenic culture (Fig. 2C). Further analysis showed a significantly increased cytoplasmic distribution of RhoA, consistent with the more prominent F-actin cytoskeleton, in cells cultured on hydrogels conjugated with Foxy5 peptide compared with those cultured on the controls. The Western blotting results showed that the expression levels of ROCK2 and p-FAK, two key mechanotransduction signaling molecules, were significantly up-regulated in the Foxy5 + RGD hydrogels (Fig. 2D) by 83 and 75% compared with those in the RGD hydrogels, respectively, and by 39 and 35% compared with those in the Scram + RGD hydrogels, respectively (Fig. 2E). Together, these data suggest that Foxy5 peptide immobilized on hydrogels is capable of initiating noncanonical Wnt signaling via the up-regulation of Dvl2, which further activates downstream RhoA signaling, leading to enhanced F-actin stability, actomyosin contractility, and cell adhesion structure development. Furthermore, the canonical Wnt signaling has been shown to promote the osteogenesis of hMSCs directly through the up-regulation of -catenin and downstream osteogenic genes including RUNX2, Dlx5, and Osterix (29). The immobilized Foxy5 peptide may indirectly facilitate the canonical Wnt signaling via the up-regulation of Frizzled3, LRP5/6, and -catenin. More thorough examinations on the effect of Wnt5a mimetic ligands on both canonical and noncanonical Wnt signaling are certainly worthy of further investigations in the future.

Our data reveal that the Foxy5 peptidemediated activation of noncanonical Wnt signaling is essential for regulating the expression and localization of critical signaling molecules involved in cell adhesion and mechanotransduction, including YAP, ROCK2, and p-FAK, which are essential for the osteogenesis of MSCs. The subtypes of ROCK, ROCK1, and ROCK2 have been shown to play distinct roles in regulating cytoskeletal tension. ROCK1 is a nonsecreted protein that destabilizes the actin cytoskeleton by regulating myosin light chain phosphorylation and peripheral actomyosin contraction, whereas ROCK2 is required for stabilizing the actin cytoskeleton by regulating cofilin phosphorylation (30). Previous studies have revealed that ROCK2 activity is effectively activated upon Wnt5a ligation to its receptors (22). We observed the up-regulation of ROCK2 expression and the down-regulation of ROCK1 expression, along with a significant increase in the focal adhesion levels in the MSCs presented with hydrogel-conjugated Foxy5 peptide. Therefore, the elevated ROCK2 activity results in enhanced cytoskeletal stability and more robust mechanotransduction signaling, both of which contribute to enhanced osteogenesis. When we inhibited ROCK activity in the Y-27632Foxy5 + RGD group using 10 M Y-27632, the expression of osteogenic genes was greatly reduced, thereby further confirming the important role of ROCK2 in Foxy5 peptideinduced osteogenesis.

Apart from RhoA activation, noncanonical Wnt5a activation has also been reported to lead to the mobilization of free intracellular calcium, which regulates multiple cellular behaviors, including the motility and differentiation of MSCs (31). To test the effect of Foxy5 peptide on the intracellular calcium level, we subjected hMSCs to Furaacetoxymethyl (AM) staining after being seeded on 2D peptide-conjugated MeHA hydrogels and cultured in osteogenic media. After 7 days of osteogenic culture, Fura-AM staining showed that cells on Foxy5 + RGD hydrogels exhibited 122 and 127% higher fluorescence intensity than those on RGD and Scram + RGD hydrogels, respectively (Fig. 2F). This finding suggests that the conjugated Foxy5 peptide is capable of activating noncanonical Wnt signaling to elevate the intracellular calcium level of MSCs, promoting osteogenesis.

Calcium-dependent noncanonical Wnt signaling pathways are known to participate in osteoblast differentiation, maturation, and bone formation (31, 32). Intracellular Ca2+ and calcium-binding/activatable signaling factors (calmodulin, calmodulin kinase II, calcineurin, etc.) are critical to the growth and differentiation of osteoblasts (33). Our biochemistry analysis and histological staining further showed that the amount of bone ECM production by hMSCs was significantly enhanced together with the intracellular calcium concentration in the hydrogels conjugated with the Foxy5 peptide, and this indicates that the elevated intracellular calcium level contributes to the enhanced osteogenesis of MSCs seeded in Foxy5 peptidefunctionalized hydrogels.

The guided lineage commitment of stem cells is a critical prerequisite for successful and efficient tissue regeneration, which is known to be modulated by the concerted actions of multiple microenvironmental signals (34). We next examined whether the hydrogels functionalized with the Wnt5a mimetic peptide could promote the osteogenic differentiation of hMSCs. We cultured hMSCs on 2D hydrogel substrates that were functionalized with RGD alone (RGD) or RGD with either Foxy5 peptide (Foxy5 + RGD) or scrambled Foxy5 peptide (Scram + RGD) in osteogenic induction media. Supplementation of the culture media with nonconjugated soluble Foxy5 peptide was previously reported to affect the chemotaxis of cancer cells (35). To compare the effects of the freely diffusing soluble form and the hydrogel-immobilized form of Foxy5 peptide on hMSCs, we included two control groups in which the osteogenic medium was supplemented with soluble, free, nonconjugated Foxy5 or scrambled Foxy5 peptide (free Foxy5 and free Scram) in the same amounts as those present in the conjugated hydrogels (Foxy5 + RGD and Scram + RGD).

Previous studies have demonstrated the critical role of YAP/TAZ-mediated mechanotransduction signaling in osteogenesis (36). We performed immunofluorescence staining for YAP and RUNX2 after 7 days of osteogenic culture. The hMSCs cultured on Foxy5 + RGD hydrogels consistently exhibited more YAP nuclear localization than those in all other control groups on 2D hydrogels (Fig. 3A). Specifically, quantification of the average nuclear-to-cytoplasmic staining intensity ratio (N/C ratio) of YAP in at least 20 representative cells from each group showed that the YAP nuclear localization in the Foxy5 + RGD group was 134 and 88% higher than that in the RGD and Scram + RGD groups, respectively (Fig. 3, A and D). Moreover, the addition of soluble Foxy5 peptide in the culture media (free Foxy5) failed to significantly increase the YAP nuclear localization as much as the hydrogel-conjugated Foxy5 peptide (only a 53% increase compared with the RGD group), whereas the free, soluble scrambled peptide had no significant effect (Fig. 3, A and C). This finding indicates that the hydrogel-conjugated Foxy5 peptide has significantly higher bioactivity than the unconjugated Wnt5a peptide directly added to the media in terms of promoting mechanosensing and osteogenesis. The immobilization of this ligand on the porous scaffold greatly enhanced the local effective concentration in the microenvironment of hMSCs, thereby facilitating the ligation of Wnt5a ligands to the receptors on the cell membrane. In contrast, the direct supplementation of the ligand resulted in its dilution in the entire volume of the media and therefore reduced the local effective ligand concentration. We also speculated that the immobilized ligands are unlikely to be internalized by cells upon ligation to membranous receptors, whereas the free ligands can be quickly internalized by cells and lose their activation function (37).

(A) Fluorescence micrographs of hMSCs stained for F-actin (red), nuclei (blue), and the mechanosensing marker YAP (green) or the osteogenic marker RUNX2 (green) (B) and ALP (blue in bright-field) (C), cultured on the RGD, Foxy5 + RGD, and Scram + RGD hydrogels. (D) Analysis of the nuclear localization of YAP determined by the nuclear-to-cytoplasmic fluorescence intensity ratio (N/C ratio) (n = 20) and (E) RUNX2 nuclear localization (n = 20) and (F) ALP expression of representative cells cultured on 2D hydrogels in the different experimental groups (n = 9). Scale bars represent 50 m in the fluorescence micrographs and 200 m in the bright-field images. Data are shown as the means SD. Statistical significance: *P < 0.05, **P < 0.01, and ***P < 0.001 significant difference.

To examine the effect of YAP nuclear accumulation on the osteogenesis of hMSCs, we analyzed the early osteogenic lineage commitment by immunofluorescence staining for RUNX2, an essential osteogenic transcription factor, after 7 days of osteoinductive culture. Cells in the Foxy5 + RGD group showed RUNX2 nuclear-to-cytoplasmic ratio 97 and 116% greater than that in the RGD and Scram + RGD groups, consistent with the YAP nuclear localization results (Fig. 3, B and E). In contrast, the expression levels of RUNX2 in the free Foxy5 and free Scram groups were only slightly higher than those in the RGD group. Furthermore, staining for ALP, another key osteogenesis marker, showed that the average percentage of ALP-positive cells in the Foxy5 + RGD group was 55, 52, 94, and 77% higher than that in the RGD, Scram + RGD, free Foxy5, and free Scram groups, respectively (Fig. 3, C and F). These findings indicate that hydrogel-conjugated Foxy5 peptide promotes the mechanosensing-dependent osteogenic differentiation of hMSCs and that the immobilization of Foxy5 peptide on hydrogels is more effective than the continuous supplementation of media with soluble Foxy5 peptide to enhance the osteogenesis of hMSCs. To further examine the effectiveness of the immobilized Foxy5 peptide on the substrates with different stiffness, we analyzed the mechanosensing and the early osteogenic lineage commitment by immunofluorescence staining for YAP and RUNX2 on the 2D MeHA hydrogels with varying stiffness of 2, 5, and 14 kPa (38). Cells in the Foxy5 + RGD group showed significantly higher YAP and RUNX2 nuclear-to-cytoplasmic ratio than that in the RGD and Scram + RGD groups at each of the selected hydrogel stiffness levels (fig. S6). This indicates that the biomaterial-conjugated Foxy5 peptide promotes osteogenesis in a wide range of substrate stiffness, and we found that the pro-osteogenic effect of the conjugated Foxy5 peptide is more significant at low substrate stiffness (2 kPa) (fig. S6).

We next examined the effect of Foxy5 peptide conjugated to 3D porous hydrogels on the osteogenic differentiation of seeded hMSCs from three different donors after 7 days (Fig. 4, A and B, fig. S7A, and tables S1 and S2) and 14 days (fig. S7B) of osteogenic culture. The real-time quantitative polymerase chain reaction (RT-qPCR) data showed that the expression levels of type I collagen, ALP, RUNX2, and OPN in cells seeded in the Foxy5 peptidefunctionalized porous hydrogels (Foxy5 + RGD group) were up-regulated by 33, 57, 35, and 422%, respectively, compared with those in cells seeded in the hydrogels without Foxy5 functionalization (RGD group) (Fig. 4B) after 7 days of osteogenic culture. In contrast, presentation of the nonfunctional scrambled peptide (Scram + RGD group) had no significant influence on the expression levels of these osteogenic genes compared with corresponding levels in the RGD group. The pro-osteogenic effect of the conjugated Foxy5 peptide diminished after 14 days of culture (fig. S7B). The diminishing effect of Foxy5 peptide can be attributed to the decreasing membrane presence of available LRP5/6 in the osteogenically differentiating hMSCs (39, 40). In addition, the increasing extracellular matrix that accumulated around hMSCs over time may have also contributed to the effect of conjugated Foxy5 peptide. It is noteworthy that the declining expression of Wnt receptors and Wnt signaling are essential for the formation of mineralized matrix (39). Therefore, this diminished effect of Foxy5 peptide over time may be beneficial to the osteogenesis of seeded hMSCs and subsequent neobone formation. Consistent with the gene expression data, both Alizarin Red and von Kossa staining revealed more substantial calcification in the Foxy5 + RGD group than in the other control groups. Quantitative analysis showed that the Alizarin Red and von Kossa staining intensities in the Foxy5 + RGD group were 262 and 107% higher than those in the RGD groups after 14 days of culture, respectively (Fig. 4, C and D). Furthermore, we assessed the organic bone matrix synthesis of stem cells by type I collagen staining. The Foxy5 + RGD group exhibited 163 and 179% higher type I collagen staining than the RGD and Scram + RGD groups, respectively (Fig. 4, C and D). These findings suggest that Foxy5 conjugated to the 3D porous hydrogel scaffold significantly promotes the expression of both early- and late-stage osteogenic genes in hMSCs and inorganic/organic bone matrix synthesis.

(A) Schematic illustration of the seeding of hMSCs in the Foxy5/Scram + RGD peptidefunctionalized 3D hydrogel scaffolds. (B) Quantitative gene expression of osteogenic markers (type I collagen, RUNX2, ALP, and OPN) in hMSCs seeded in porous hydrogels conjugated with RGD peptide alone (RGD), Foxy5 and RGD peptides (Foxy5 + RGD), or Scram and RGD peptides (Scram + RGD) in osteogenic culture. (C) von Kossa staining, Alizarin Red S staining, and immunohistochemistry staining of type I collagen and (D) quantification of the staining intensities after 14 days of osteogenic culture. Scale bars, 50 m. Data are shown as the means SD (n = 9). Statistical significance: * P < 0.05, **P < 0.01, and ***P < 0.001.

In mature bone tissues, the unique osteoblastic microenvironment niche provides MSCs with the necessary biological cues, including stromal cellderived factor 1, angiopoietin 1, and OPN, derived from osteoblasts, osteoclasts, osteocytes, and endothelial cells in trabecular bone and bone marrow (19, 41). Many previous studies have reported the regulation of MSC signaling and lineage commitment by systemic hormones or localized growth factors (42). Fu et al. demonstrated that Wnt3a-mediated canonical Wnt signaling activation antagonizes the terminal osteogenic differentiation of MSCs, while Wnt5a-mediated noncanonical Wnt signaling mitigates the inhibitory effect of Wnt3a. In the natural osteoblastic niche, Wnt5a proteins are secreted by the surrounding tissues and bind to the Frizzled/Ror2 surface Wnt receptors of hMSCs via paracrine mechanisms (43). We used porous hydrogels functionalized with Wnt5a mimetic Foxy5 peptide to emulate this pro-osteogenic niche and promote the osteogenesis of hMSCs. In addition to Foxy5 peptide, RGD peptide was also conjugated to all hydrogels to provide adhesive motifs for the hMSCs because Foxy5 peptide alone cannot support effective cell adhesion. The promechanotransduction and pro-osteogenic effects of Foxy5 peptide were therefore not studied in the absence of RGD peptides, which is a limitation of the study. Nevertheless, integrin ligands are important components of the natural osteogenic niche in bones. The potential crosstalk between integrin signaling and Wnt signaling and the associated effects on stem cell differentiation certainly warrant further investigation.

We further evaluated the efficacy of the Foxy5 peptide conjugated to the hydrogel in assisting in vivo bone regeneration in rat calvarial defects. Rat MSCs (rMSCs) with trilineage differentiation potential (4446) were first seeded in porous hydrogels functionalized with RGD, Foxy5 + RGD, or Scram + RGD peptides and cultured in osteogenic media for 7 days prior to transplantation into the defects (Fig. 5A). The RT-qPCR data showed that the expression levels of type I collagen, ALP, RUNX2, OPN, Dvl2, RhoA, and vinculin in rMSCs of the Foxy5 + RGD group were all significantly higher compared with those in the control groups (RGD group, Scram + RGD) after 7 days of osteogenic culture (fig. S8, A and B). Eight weeks after transplantation, both hematoxylin and eosin (H&E) staining and immunohistochemical staining for osteocalcin and type I collagen revealed enhanced osteoblastic marker expression and bone matrix formation in the Foxy5 + RGD group compared with the RGD, Scram + RGD, and blank groups (Fig. 5B). The average staining intensity for osteocalcin was 124, 114, and 240% higher and that of type I collagen was 105, 104, and 144% higher in the Foxy5 + RGD group than in the blank, RGD, and Scram + RGD groups, respectively (Fig. 5D). The average staining intensity for osteocalcin and type I collagen in the Foxy5 + RGD group was still around 30% lower than that in the native calvarial tissue (Fig. 5D). Furthermore, the microcomputed tomography (micro-CT) reconstruction data revealed considerably more new bone formation in the defects treated with hydrogels conjugated with Foxy5 and RGD peptides (Foxy5 + RGD group) than in those treated with the control hydrogels (blank, RGD, and Scram + RGD groups) (Fig. 5C). Quantitative analysis showed that the bone volumetototal tissue volume (BV/TV) ratio in the Foxy5 + RGD group was 107, 57, and 198% higher than that in the RGD, Scram + RGD, and sham blank groups, respectively. Notably, the average BV/TV ratio of healthy calvarial bone was determined to be 35.25% due to other skeletal components, such as fibrous connective tissues. The average BV/TV ratio in the Foxy5 + RGD group was 25.19%, suggesting a substantial recovery of approximately 71.49% of the healthy calvarial bone volume. These findings demonstrate that the functionalization of biomaterial scaffolds with this Wnt5a mimetic peptide can substantially enhance bone regeneration in vivo. No significant abnormalities in tissue structure or morphology were observed around the implantation site. We believe that the restricted bioactivity of the mimetic peptide (compared with that of the parent protein) and the immobilization of this Wnt5a ligand on the biomaterial for local implantation will limit potential undesired nonspecific actions that may arise due to peptide transportation to other off-target sites (47).

(A) Schematic illustration of the implantation of rMSC-seeded and peptide-functionalized porous hydrogels in rat calvarial defects. (B) H&E staining and immunohistochemical staining of the native healthy bone tissue and the calvarial defects treated with the RGD hydrogels, Foxy5 + RGD hydrogels, Scram + RGD hydrogels, and no hydrogels (blank) 8 weeks after implantation (n = 3). High-magnification images showing the defect/native bone boundaries highlighted in yellow and red boxes and defect center areas in blue boxes in the low-magnification images of H&E-stained sections. The dotted lines indicate the boundary between the defect and native bone. The newly formed bone was seamlessly integrated with the neighboring native bone in the Foxy5 + RGD group. Scale bars, 50 m. (C) Top view of 3D micro-CT images showing calvarial bone defects after 8 weeks in all groups (n = 3). (D) Bone volume (normalized to total tissue volume, BV/TV) in the calvarial defects in all groups after 8 weeks (n = 3). The bone volume of healthy rat calvarial bone is shown as the benchmark. Quantification of the immunohistochemical staining intensity of the osteogenic markers, including osteocalcin and type I collagen, showing the higher intensity in the Foxy5 + RGD group compared with those of the RGD and Scram + RGD control groups. Data are shown as the means SD (n = 9). Statistical significance: *P < 0.05, **P < 0.01, and ***P < 0.001 significant difference.

MeHA macromolecules were synthesized from sodium hyaluronate powder (molecular weight, ~74 kDa; Lifecore, Chaska, Minnesota, USA), as previously reported (8). Briefly, 100 ml of 1% (w/v) sodium hyaluronate solution was reacted for 24 hours with 4 or 1.5 ml of methacrylic anhydride at pH 9.5, adjusted with 2 M NaOH solution. After complete dialysis and lyophilization, 100% methacrylation or 30% methacrylation was confirmed using proton nuclear magnetic resonance (1H NMR). The RGD peptide (GCGYGRGDSPG) and Foxy5 peptide (MDGCEL) (GenScript, Nanjing, Jiangsu, China) with a cysteine amino acid at the C-terminal end were conjugated to the MeHA backbone with a Michael addition reaction between the methacrylate groups and the thiol groups of each peptide in basic phosphate buffer (pH 8.0) containing 10 M tris(2-carboxyethyl)phosphine at 37C. The molar ratio of methacrylate to each peptide thiol was 100:3. RGD-functionalized, Foxy5-conjugated porous MeHA hydrogels were fabricated from 50 l of the peptide-conjugated MeHA solution (3% w/v, 100% methacrylation) after 2 hours, with 1.51 mol of DTT as the cross-linker to consume all residual methacrylic groups, in round polyvinyl chloride molds fully packed with a poly(methyl methacrylate) (PMMA) microsphere porogen ( 200 m). The constructs generated were immersed in acetone and shaken at 90 rpm to dissolve the PMMA porogen, sterilized with 75% ethanol for 1 day, and rinsed three times with sterile phosphate-buffered saline (PBS). In the directly Foxy5 peptidesupplemented MeHA (free Foxy5) group and the directly scrambled peptidesupplemented MeHA (free Scram) group, we only used the same Foxy5 + RGDfunctionalized MeHA solution or Scram + RGDfunctionalized MeHA solution (3% w/v, 100% methacrylation) to fabricate the porous hydrogels, respectively.

We used MeHA with 30% methacrylation supplemented with 0.05% (w/v) photoinitiator I2959 (Sigma-Aldrich, MO, USA) to make Foxy5 + RGDfunctionalized MeHA solution, Scram + RGDfunctionalized MeHA solution, or RGD-functionalized MeHA solution (3% w/v, 30% methacrylation) and to fabricate 2D hydrogels with different stiffness in polyvinyl chloride molds under 367, 467, 667, or 1067 seconds of UV exposure. All 2D hydrogels were sterilized before cell culture. The surface roughness and modulus were determined by atomic force microscopy (Bruker, MA, USA).

2D-biofunctionalized substrates were fabricated by polymerizing peptide-conjugated MeHA precursor solutions (3% w/v, 30% methacrylation) under UV light (wavelength, 365 nm; intensity, 7 mW/cm2) on methacrylated glass coverslips. The porous MeHA hydrogel constructs (height, ~1.5 mm; 1 mm) were polymerized in molds filled with 200-m PMMA microbeads to form an interconnected porous structure for in vitro experiments and for in vivo calvarial defect regeneration (Fig. 1, B and C) (9). Homogenous, interconnected spherical structures within the hydrogels were characterized through bright-field images captured using a fluorescence microscope (Nikon, Japan) (Fig. 1D). The Young moduli of the hydrogels were determined using a mechanical tester (Mach-1, Biomomentum Inc., Suite, Canada), and the strain-controlled frequency sweep mechanical tests were performed using a rheometer (Malvern Inc., Malvern, Britain).

Passage-4 hMSCs (Lonza, Walkersville, Maryland, USA) were expanded in basal growth medium [-minimal essential medium (MEM) supplemented with 16.7% (v/v) fetal bovine serum (FBS), penicillin-streptomycin (P/S; 100 U ml1), and 2 mM l-glutamine]. Growth medium (50 l) containing 5 105 hMSCs (108 cells ml1) was injected into one semidry, porous MeHA-RGD hydrogel, which was incubated at 37C for 4 hours to allow cell attachment to the hydrogels. Then, 1 ml of osteogenic medium [-MEM, 16.67% FBS, 1% P/S, 2 mM l-glutamine, 10 mM -glycerophosphate disodium, l-ascorbic acid 2-phosphate (50 mg ml1), and 100 nM dexamethasone] was added to all the hydrogels, and the medium was changed every 2 days. In the control group, Foxy5 peptide or scrambled peptide solution was added directly to the hydrogels at the beginning of osteogenic culture. Samples were collected on days 7 and 14 to evaluate the degree of osteogenesis by traditional qPCR and immunofluorescence staining. Cell viability was determined by alamarBlue assay (Invitrogen, Carlsbad, California, USA) after 7 days in osteogenic culture. Live/dead staining was performed by adding 3 M calcein AM and 3 M propidium iodide (Thermo Fisher Scientific, Waltham, Massachusetts, USA) to the hydrogels. After incubation for 30 min at 37C, the hydrogels were washed three times with PBS, and fluorescence images were captured using a confocal microscope (Nikon C2, Tokyo, Japan). The quantification of immunofluorescence staining results was conducted by using the ImageJ software [National Institutes of Health (NIH), Baltimore, Maryland, USA]. First, we adjusted the color images of the immunofluorescence staining to the 8-bit grayscale images, and then we selected the region of cells that best represent the overall staining intensity of the samples in the immunofluorescence staining results. The average grayscale values of the region of interest taken from at least 20 cells in each group were determined and compared to get the quantification results. The reported data of biochemical assays are the pooled results from three experiments.

All samples were homogenized in 1 ml of TRIzol reagent (Invitrogen), and total RNA was extracted according to the manufacturers protocol. The RNA concentration was measured using a NanoDrop One spectrophotometer (NanoDrop Technologies, Waltham, Massachusetts, USA). Total RNA (1 g) was reverse transcribed into cDNA using the RevertAid First Strand cDNA Synthesis Kit (Thermo Fisher Scientific). qPCR was performed on an Applied Biosystems StepOnePlus Real-Time PCR System with TaqMan primers and probes (Applied Biosciences, Waltham, Massachusetts, USA) specific for glyceraldehyde 3-phosphate dehydrogenase (GAPDH) and other osteogenic genes, including those encoding RUNX2, ALP, and type I collagen. The sequences of the TaqMan primers and probes used are listed in table S2. The osteogenic gene expression levels were normalized to that of GAPDH, and the relative expression levels were calculated with the 2Ct method.

Hydrogel samples were fixed overnight in 4% paraformaldehyde at 4C, dehydrated in a graded series of ethanol, crystalized in a graded series of xylene, and embedded in paraffin. Histological sections (7 m) were stained for type I collagen using the VECTASTAIN ABC Kit and the DAB Substrate Kit (Vector Laboratories, Burlingame, California, USA). Briefly, hyaluronidase (0.5 g liter1) was applied to the rehydrated samples to predigest them at 37C for 30 min. The samples were then incubated with 0.5 N acetic acid for 4 hours at 4C to induce swelling, followed by incubation at 4C overnight with a primary antibody directed against type I collagen (antitype I collagen, diluted 1:200; sc-59772, Santa Cruz Biotechnology). Immunofluorescence staining for the osteogenic-related proteins (YAP and RUNX2) and cellular contractionrelated proteins (RhoA) was conducted as previously reported (9). The calcification of phosphates and calcium ions was identified with von Kossa staining and Alizarin Red S staining, respectively. Briefly, von Kossa stain was applied to the rehydrated sections, as previously reported. To stain with Alizarin Red S, 1 ml of 0.5% (w/v) Alizarin Red S solution (Sigma) was applied to each hydrogel sample; the samples were then incubated for 5 min at room temperature before being washed, dehydrated, cleared, and sealed. Images were captured using a bright-field microscope (Nikon). The quantification was conducted by using the ImageJ software (NIH). First, we adjusted the color images of immunohistochemistry (IHC) staining to the 8-bit grayscale images, and then we selected regions of interests of identical size that best represent the overall staining intensity of the samples in the IHC staining results. The average grayscale values of the region of interest taken from three parallel samples in each group were determined and compared to get the quantification results. The reported data of biochemical assays are pooled results from three experiments.

Strictly following the guidelines of the Institutional Animal Care and Use Committee at The Chinese University of Hong Kong, 12-week-old male Sprague-Dawley rats were randomly divided into four groups, shaved, and prepped for aseptic surgery. A midline skin flap was raised over the parietal bones and reflected caudally to expose the midsagittal and transverse sutures. The periosteum was incised along the midsagittal suture and the right or left transverse suture and removed to expose the parietal bone. A 5-mm-diameter defect was created using a trephine with normal saline irrigation during processing, and a section of the bone was removed to expose the dura mater. 3D porous peptidefunctionalized HA hydrogels (n = 3 per group) 5 mm in diameter and 1 mm thick were seeded with 1 million rMSCs with trilineage differentiation potential (46). After 7 days of in vitro osteogenic induction in the incubator, the hydrogels were implanted into the calvarial defects. All experimental animals were maintained until 8 weeks from the day of defect creation. After the rats were euthanized, the parietal bones were harvested and decalcified with 10% EDTA solution. The subsequent H&E staining and histological analysis were performed as described in previous publications (9).

All data are presented as the means SD. Statistica (Statsoft, Tulsa, Oklahoma, USA) was used to perform the statistical analyses using two-way analysis variance (ANOVA) and Tukeys honest significant difference post hoc test of the means; the culture period and experimental groups were used as independent variables.

Acknowledgments: We are grateful for the technical support from J. Lai, S. Wong, and A. Cheung from the School of Biomedical Sciences (The Chinese University of Hong Kong). We thank M. Zhu and K. Zhang for proofreading the manuscript. We sincerely thank M. Wong, N. So, K. Wei, M. Zhu, B. Yang, H. Chen, K. Zou, K. Zhang, Y. Jing, D. Siu Hong Wong, X. Xu, E. Yingrui Deng, X. Chen, and W. Li for the valuable discussions, support, and love. Funding: Project 31570979 is supported by the National Natural Science Foundation of China. The work described in this paper is supported by a General Research Fund grant from the Research Grants Council of Hong Kong (project nos. 14202215 and 14220716). This research is also supported by project BME-p3-15 of the Shun Hing Institute of Advanced Engineering (The Chinese University of Hong Kong). This work is supported by the Health and Medical Research Fund, the Food and Health Bureau, the Government of the Hong Kong Special Administrative Region (reference no. 04152836). This research is supported by the Chow Yuk Ho Technology Centre for Innovative Medicine (The Chinese University of Hong Kong). The work was partially supported by the Hong Kong Research Grants Council Theme-based Research Scheme (reference no. T13-402/17-N, Functional Bone Regeneration in Challenging Bone Disorders and Defects, 1 November 2017 to 31 October 2022). Author contributions: S.L. contributed to the animal experiments and analysis. M.Z. and J.X. contributed to the peptide synthesis and porous gel fabrication. Y.D. contributed to the cell culture and qPCR assay. X.C. contributed to the polymer synthesis and NMR characterization. K.W. contributed to the macromer synthesis and proofreading of the manuscript. R.L. contributed to the rest of the experiments and the manuscript. G.L. led the in vivo study of the project. L.B. led the project as the supervisor. Competing interests: The authors declare that they have no competing interests. Data and materials availability: All data needed to evaluate the conclusions in the paper are present in the paper and/or the Supplementary Materials. Additional data related to this paper may be requested from the authors.

Here is the original post:
Synthetic presentation of noncanonical Wnt5a motif promotes mechanosensing-dependent differentiation of stem cells and regeneration - Science Advances

The Prognostic Importance of EVI1 Expression – Cancer Network

In AML, the role of ecotropic virus integration site-1 (EVI1) expression is debated. To date, results of studies have been mixed with only some studies demonstrating EVI1 expression related to poorer survival. In a meta-analysis published in the Annals of Hematology, researchers set out to uncover the predictive capability of this marker.

As a malignant disorder in hematology usually with a poor prognosis, AML needs an accurate prediction of prognosis to indicate protocoling the appropriate therapy regimens for patients hoping for survival improvement, wrote authors, led by Xia Wu, Department of Hematology, West China Hospital, Sichuan University, Sichuan Province. Molecular markers increasingly play an utmost significant role in the diagnosis and risk stratification of AML.

In the current meta-analysis, Wu et al. mined 11 studies for 4767 AML patients with intermediate cytogenetic risk (ICR), according to National Comprehensive Cancer Network (NCCN), International System for Human Cytogenetic Nomenclature (ISCN), or European leukemia network (ELN) guidance. The findings indicated that EVI1 expression negatively impacted OS (HR = 1.73, 95%CI 1.432.11) and event=free survival or EFS (HR = 1.17, 95%CI 1.051.31). Furthermore, EVI1 was a negative predictor of prognosis in patients with normal cytogenetics (NC) and younger patients (< 60 years).

Importantly, the investigators noted that due to location, altered EVI1 most often accompanies 3q26 rearrangements. However, it remains to be elucidated whether increased EVI1 expression is related to AML outcomes in those without 3q mutations. On a related note, higher levels of EVI1 may affect AML subgroups differently, which, according to the authors, is of utmost significance for clinical physicians.

In other findings, EVI1H expression was rarely found with NPM1, FLT3-ITD and DNMT3A mutations. Wu et al point to these mutations and mutations as avenues of further research.EVI1 is a transcription factor on chromosome 3. It was first discovered two decades ago in murine models. It has stem cell specific expression patterns and mediates growth of hematopoietic stem cells, and plays a role in AML, myelodysplastic syndrome (MDS), and CML.

The investigators suggested that the findings of the current study could assist clinicians with risk stratification and treatment decisionsespecially because most patients are NC.

EVI1, which also goes by MECOM, encodes a 145 kDa-unique zinc finger that attaches with DNA. This transcription factor is hypothesized to interfere with granulocyte and erythroid cell differentiation, as well as promotion of megakaryocyte breakdown, to aid with the differentiation and proliferation of hematopoetic stem cells.Several drug targets for EVI1 have been suggested such those involved in leukemogenesis and stem cell maintenance. Examples include the transcription factor Pre-B Cell leukemia Homeobox 1 (PBX1) and Phosphatase and Tensin Homolog (PTEN), which is a tumor suppressor gene. However, none of these targets have proven related to EVI1-deregulated AML.

Per the authors the current study had several limitations. First, most studies in the meta-analysis were observational and not randomized-controlled trials. Second, the sample contained cases of therapy-related AML and secondary AML, which have a worse prognosis and could thus confound results. Third, limited OS data precluded the ability to study AML patients without 3q alterations. Fourth, the studies were highly heterogeneous in a clinical sense.

Reference

Wu X et al. Prognostic significance of the EVI1 gene expression in patients with acute myeloid leukemia: a meta-analysis. Annals of Hematology. 2019 Sep 3. doi: 10.1007/s00277-019-03774-z.

View post:
The Prognostic Importance of EVI1 Expression - Cancer Network