Author Archives: admin


Global Cell Analysis Instruments Market (2020 to 2025) – Growth, Trends, and Forecasts – Yahoo Finance UK

Dublin, Aug. 05, 2020 (GLOBE NEWSWIRE) -- The "Cell Analysis Instruments Market - Growth, Trends, and Forecasts (2020 - 2025)" report has been added to ResearchAndMarkets.com's offering.

The cell analysis instruments market is found increasing due to the growing burden of cancer across the globe and growing biotech industry and research in cell biology.

Cancer is one of the major causes of deaths across the world, with 609,640 victims in 2018, accounting for nearly one in six deaths. The disease has witnessed significant growth in the last few decades and is expected to rise rapidly over the forecast period. According to the National Cancer Institute, there were around 1,735,350 cases of cancer found in the United States during the year 2018.

In addition, WHO has estimated the cancer deaths to increase by 70% over the next two decades, worldwide. The recent years have observed significant inclination by biotech research companies and universities to invest in various single-cell DNA and RNA quantification techniques. Hence, all these factors are found increasing the demand for cell analysis instruments market.

Key Market Trends

Cell Counting Application is Expected to Register a High Growth During the Forecast Period

Cell counting is done to compare different cell populations or responses. It is the fundamental and critical to numerous biological experiments. Cell counting is done by various methods such as automated cell counter, cell cycle analysis, counting cells without cell staining. The cell counting is one of the most important steps in biology and medicine. The cell counting used in medicine to understand the concentration of blood cells which can provide crucial information about the health situation of a person. In cell therapy, it is used to control the dose of cells given to the patient.

Similarly, cell counting is also used to know the concentration of bacteria, viruses and other pathogens in the blood or in other bodily fluids can reveal information about the progress of infectious disease and about the degree of success with which the immune system is dealing with the infection. With the wide applications of cell counting, it is expected that this segment will be witnessing a good CAGR over the forecast period. In addition, increasing burden of life-threatening diseases such as cancer is augmenting the growth of market studied.

North America is Expected to Dominate the Cell Analysis Instruments Market

North America is expected to dominate the market due to the increasing incidences of cancer and increasing corporate & government funding in cell-based research. Being the most developed economy and healthcare industry, the United States has put a benchmark for several markets. Due to the presence of major companies and the strong R&D in biotechnology, the country is witnessing rapid growth in the market studied. The focus of the US government is to further strengthen its healthcare industry, thus there has been a significant increase in the research funding.

Along with that, the country has a large number of laboratories which is augmenting the demand for cell analysis instruments, and ultimately, the US holds the largest share of the market owing to the high demand.

Competitive Landscape

The market for cell analysis instruments is moderately competitive and consists of local as well as global players. Developing countries are having a large number of local players that are catering to consumers across laboratories and academia. However, the largest share of the market on the global scale is grabbed by international companies such as Thermo Fisher Scientific, Sartorius AG, Danaher Corporation, etc. With the advancements being done in the biotech industry, it is believed that there will be an emergence of the new players in the future.

Reasons to Purchase this report:

Story continues

Key Topics Covered:

1 INTRODUCTION 1.1 Study Deliverables 1.2 Study Assumptions 1.3 Scope of the Study

2 RESEARCH METHODOLOGY

3 EXECUTIVE SUMMARY

4 MARKET DYNAMICS 4.1 Market Overview 4.2 Market Drivers 4.2.1 Increasing Cancer Incidence Rate 4.2.2 Growing Biotech Industry and Research in Cell Biology 4.3 Market Restraints 4.3.1 High Cost of Cell Analysis 4.3.2 Complex Regulatory Scenario 4.4 Porter's Five Force Analysis 4.4.1 Threat of New Entrants 4.4.2 Bargaining Power of Buyers/Consumers 4.4.3 Bargaining Power of Suppliers 4.4.4 Threat of Substitute Products 4.4.5 Intensity of Competitive Rivalry

5 MARKET SEGMENTATION 5.1 By Product 5.1.1 Instruments 5.1.1.1 Microscopes 5.1.1.2 Flow Cytometers 5.1.1.3 Spectrophotometers 5.1.1.4 Cell Microarrays 5.1.1.5 Others 5.1.2 Consumables 5.2 By Application 5.2.1 Cell Counting 5.2.2 Cell Viability 5.2.3 Cell Identification 5.2.4 Target Identification 5.2.5 PCR 5.2.6 Others 5.3 By End-User 5.3.1 Academic and Research Institutes 5.3.2 Pharmaceutical & Biotechnology Companies 5.3.3 Others 5.4 Geography 5.4.1 North America 5.4.1.1 United States 5.4.1.2 Canada 5.4.1.3 Mexico 5.4.2 Europe 5.4.2.1 Germany 5.4.2.2 United Kingdom 5.4.2.3 France 5.4.2.4 Italy 5.4.2.5 Spain 5.4.2.6 Rest of Europe 5.4.3 Asia-Pacific 5.4.3.1 China 5.4.3.2 Japan 5.4.3.3 India 5.4.3.4 Australia 5.4.3.5 South Korea 5.4.3.6 Rest of Asia-Pacific 5.4.4 Middle-East and Africa 5.4.4.1 GCC 5.4.4.2 South Africa 5.4.4.3 Rest of Middle-East and Africa 5.4.5 South America 5.4.5.1 Brazil 5.4.5.2 Argentina 5.4.5.3 Rest of South America

6 COMPETITIVE LANDSCAPE 6.1 Company Profiles 6.1.1 Becton Dickinson and Company 6.1.2 Sartorius AG 6.1.3 Danaher Corporation 6.1.4 Merck KGaA 6.1.5 Thermo Fisher Scientific 6.1.6 Miltenyi Biotec 6.1.7 STEMCELL Technologies 6.1.8 Terumo BCT 6.1.9 Promega Corporation 6.1.10 GE Healthcare

7 MARKET OPPORTUNITIES AND FUTURE TRENDS

For more information about this report visit https://www.researchandmarkets.com/r/rmp0ll

Research and Markets also offers Custom Research services providing focused, comprehensive and tailored research.

View original post here:
Global Cell Analysis Instruments Market (2020 to 2025) - Growth, Trends, and Forecasts - Yahoo Finance UK

Edited Transcript of XNCR.OQ earnings conference call or presentation 4-Aug-20 8:30pm GMT – Yahoo Finance

Monrovia Aug 5, 2020 (Thomson StreetEvents) -- Edited Transcript of Xencor Inc earnings conference call or presentation Tuesday, August 4, 2020 at 8:30:00pm GMT

Xencor, Inc. - Senior VP & Chief Medical Officer

Xencor, Inc. - Co-Founder, CEO, President & Director

Xencor, Inc. - Associate Director and Head of Corporate Communications & IR

Xencor, Inc. - Senior VP & CFO

Xencor, Inc. - Senior VP of Research & Chief Scientific Officer

SVB Leerink LLC, Research Division - MD of Emerging Oncology & Senior Research Analyst

Joh. Berenberg, Gossler & Co. KG, Research Division - Analyst

Good afternoon, ladies and gentlemen, and thank you for standing by, and welcome to the Second Quarter 2020 Xencor Conference Call. (Operator Instructions)

Now I would like to turn the call to your speaker today, Charles Liles, Head of Investor Relations.

Charles Liles, Xencor, Inc. - Associate Director and Head of Corporate Communications & IR [2]

Thank you, and good afternoon.

Earlier today, we issued a press release which outlines the topics we plan to discuss today. The press release is available at http://www.xencor.com. Today on our call, Bassil Dahiyat, President and Chief Executive Officer, will provide updates regarding COVID-19 and our partnerships; Allen Yang, Chief Medical Officer, will review recently presented clinical data; John Desjarlais, Chief Scientific Officer, will provide updates from preclinical development; and John Kuch, Chief Financial Officer, will review financial results. And then we will open up the call for your questions.

Before we begin, I would like to remind you that, during the course of this conference call, Xencor management may make forward-looking statements, including statements regarding the company's future financial and operating results. Future market conditions, the plans and objectives of management for future operations, the company's partnering efforts, capital requirements, future product offerings, research and development programs and the impacts of the COVID-19 pandemic on these topics. These forward-looking statements are not historical facts, but rather are based on our current expectations and beliefs and are based on information currently available to us. The outcome of the events described in these forward-looking statements are subject to known and unknown risks, uncertainties and other factors that could cause actual results to differ materially from the results anticipated by these forward-looking statements including, but not limited to, those factors contained in the Risk Factors section of our most recently filed annual report on Form 10-K and quarterly report on Form 10-Q.

With that, let me pass the call over to Bassil.

--------------------------------------------------------------------------------

Bassil I. Dahiyat, Xencor, Inc. - Co-Founder, CEO, President & Director [3]

--------------------------------------------------------------------------------

Thanks, Charles, and good afternoon, everyone.

Xencor's approach to creating antibody and cytokine therapeutics is centered around our XmAb protein engineering platform. By making small changes to an antibody's structure, specifically in its Fc domain, we can improve its natural functions and performance and create new mechanisms of therapeutic action. The plug-and-play nature of our suite of XmAb Fc domains allows us to engineer nearly any antibody to have improved activity, longer half-life or bispecific structure. This flexibility and portability enables us to take multiple shots on goal simultaneously in the clinic and generate proof-of-concept data to guide which programs will independently advance, which will partner and which will terminate.

We're focusing our R&D on the expansion and use of our XmAb bispecific platform to create antibodies that bind 2 or more different targets simultaneously and also to engineer cytokines with structures optimized for particular therapeutic use. Now we're currently running 6 Phase I clinical studies evaluating such XmAb bispecific antibodies.

Now before I update on some of our partnerships, we want to provide a brief update on the impact of the COVID-19 pandemic on our operation. The pandemic did not significantly disrupt patient enrollment to Xencor's 6 ongoing clinical trials during the second quarter. However, our study initiations for vibecotamab, as we've previously disclosed, have been delayed as many clinical sites have delayed the trial start-up process.

We had modestly slowed enrollment in the CD3 bispecific antibody studies attributable to COVID-19 and no effect on our studies for the 3 tumor microenvironment activator molecules. Now as is still the case today as it was 3 months ago, unfortunately, the situation is very fluid and we'll continue to update it as soon as appropriate.

Now within the company, we've implemented a number of measures to protect the health and safety of our employees and of our community. These include some laboratory operation adjustments, symptom self-assessment guidelines and weekly SARS-CoV-2 virus testing at our facility. We are maintaining a requirement for all non-laboratory employees to work remotely.

Okay. Now on to partnerships. A core part of our business is to complement our internal portfolio of -- internal development portfolio with partnering. These partnerships generate payments from the licensing of XmAb technologies, the clinical advancing of XmAb candidates as well as royalties from sales of approved products. There were no COVID-19 impacts here during the second quarter as we continue to earn revenues from partners like Alexion and Gilead, but we will continue to monitor potential impacts, of course.

Partnerships like these really highlight the plug-and-play nature of the suite of XmAb Fc domains we've created. With the small changes to the Fc structure that we've engineered, we can for nearly any antibody improve the activity, half-life or readily create bispecific structures. We have 11 ongoing partnerships for XmAb technology, which have resulted now in 2 marketed products, 7 clinical stage candidates and more in the earlier stages of development. The most significant recent development among our partners occurred just this past Friday. With the early FDA approval of MorphoSys' tafasitamab, which they licensed from us in 2010, when it was known as XmAb 5574. It's an antibody that we created and put our XmAb cytotoxic Fc domain onto. We also initiated its clinical development, running the Phase I trial. It's trade name is now Monjuvi. It's a CD19-directed cytolytic antibody indicated in combination with lenalidomide for the treatment of adult patients with relapsed or refractory diffused large B-cell lymphoma, not otherwise specified, including DLBCL arising from low-grade lymphoma and who are not eligible for autologous stem cell transplant. This approval is the first for second-line treatment of DLBCL from the FDA.

Now we couldn't be happier here at Xencor as this approval expands the options for patients with this difficult-to-treat blood cancer. Monjuvi will be co-commercialized in the U.S. by MorphoSys and Incyte, and the European marketing authorization application for tafasitamab is currently under review by the EMA.

Now from time to time, we enter into research collaborations that include the creation of novel XmAb bispecific antibodies to be advanced by partners. Amgen's a prime example. AMG 509 is Amgen's STEAP1 x CD3 XmAb 2+1 bispecific antibody. Now that was developed under our collaboration with them. They're developing AMG 509 for patients with prostate cancer and Ewing sarcoma, and a Phase I study is currently recruiting for patients with advanced prostate cancer.

Now the first bispecific antibody that Amgen developed under this collaboration is AMG 424, a CD38 x CD3 bispecific antibody that they evaluated in a Phase I study in patients with multiple myeloma. Amgen terminated the program in the second quarter and indicated the program was stopped for adverse events that were likely CD38 target-related. Under the terms of the agreement, the rights to the CD38 program, including AMG 424, revert to Xencor and the company is currently assessing the asset's potential for further development, including treating different patient populations and applying mitigating treatments for the adverse events.

Now the plug-and-play nature of our XmAb technologies enables additional partners like Alexion and Vir to advance their programs, needing very little resources directly from us. Our strategy is to selectively license access to our XmAb technologies for creating and developing antibodies with improved properties. Alexion's ULTOMIRIS, a C5 complement inhibitor uses Xtend technology for longer half-life. The program continues to receive marketing authorizations worldwide, the last of which was the European approval for adult and children with atypical hemolytic uremic syndrome, this June. In addition to evaluating ULTOMIRIS in a broad late stage development program, Alexion is currently conducting a randomized controlled Phase III study in hospitalized patients with advanced COVID-19.

Our partnership with Vir Biotech shows the broad applicability of our technology in areas such as viral infectious disease. Vir has non-exclusive access to our Xtend Fc technology to extend the half-life of VIR-7831 and VIR-7832, both novel antibodies that they're investigating as potential treatments for patients with COVID-19. They plan to submit an IND for VIR-7831 and commence a Phase II/III clinical trial program in August, and they plan to initiate a study evaluating VIR-7832 later this year.

I'll now turn it over to John Desjarlais, who will provide an update on some of our preclinical programs and our new discovery and development collaboration with Atreca. John?

--------------------------------------------------------------------------------

John R. Desjarlais, Xencor, Inc. - Senior VP of Research & Chief Scientific Officer [4]

--------------------------------------------------------------------------------

Yes. Thanks, Bassil.

Yes, and Xencor's XmAb bispecific Fc domains were specifically created to enable the rapid design and simplified development of bispecific antibodies to combine 2 or more different targets. First-in-class that we have developed were CD3 bispecific antibodies that contain 1 anti-tumor binding domain and 1 CD3 binding domain. Engagement of CD3 on T cells promotes recruitment and activation of T cells against the tumor cells. The activator receptor on T cells doesn't have to be limited to CD3, though. For example, we are also investigating bispecific antibodies that target CD28, a key co-stimulatory receptor on T cells. Importantly, we designed these CD28 engagers to activate only when bound to tumor cells, with the goal of avoiding the superagonism that led to the disastrous clinical experience of other companies targeting CD28 nearly 15 years ago.

More near term, however, we have developed a mixed valency format, our XmAb 2+1 bispecific antibody with 2 domains that bind a tumor target and a single domain that binds CD3. These antibodies may preferentially kill tumor cells with high target expression and may potentially avoid low-expressing normal cells, taking advantage of a property called avidity. We believe these properties will be particularly important for many solid tumor targets.

We presented preclinical data from 3 internally developed 2+1 bispecifics at the second session of the AACR meeting in late June. Preclinical models show strong selective tumor killing from 2+1 programs that target PSMA mesothelin and ENPP3, the last of which is an underexplored tumor antigen overexpressed on renal cell carcinomas. These targets, although they tend to be strongly expressed on prostate cancer, ovarian cancer and kidney cancer, respectively, can also have some normal tissue expression, suggesting there are good applications for this new format.

The ENPP3 program, XmAb30819, is the most advanced of these. Preclinical data indicate that XmAb30819 binds preferentially to tumor cells compared to normal cells and effectively recruits T cells to kill tumor cells selectively. It demonstrates strong reversal tumor growth in tumor xenograft models, and it was well tolerated with expected pharmacodynamics in an antibody-like half-life in nonhuman primates. We are planning to file an IND for XmAb30819 in 2021.

Finally, last month, we formalized a collaboration with Atreca to research, develop and commercialize CD3 engaging bispecific antibodies to novel targets. Atreca's unique discovery platform complements our protein engineering capabilities by providing novel tumor-selective antibodies and targets to couple with our CD3 bispecific platform. Up to 2 joint programs will be mutually selected for further development and commercialization with each partner sharing costs and profits equally. Each company will lead one of the joint programs. The agreement also allows for each partner to pursue up to 2 programs independently. This collaboration offers both Xencor and Atreca with several opportunities to advance novel first-in-class CD3 bispecific antibodies for the potential treatment of patients with cancer.

With that, Allen Yang will review our clinical portfolio. Allen?

--------------------------------------------------------------------------------

Allen S. Yang, Xencor, Inc. - Senior VP & Chief Medical Officer [5]

--------------------------------------------------------------------------------

Thanks, John.

In May, we provided initial dose escalation data from our ongoing Phase I study evaluating XmAb20717 in patients with advanced solid tumors. XmAb20717 is a dual PD-1, CTLA-4 checkpoint-inhibiting bispecific antibody. We have tuned the antibodies affinities for PD-1 and CTLA-4 for selective engagement of T cells expressing both targets, which distinguishes it from combination therapy and most bispecific checkpoint inhibitors.

T cells that have multiple checkpoint expression are typically found more in the tumor microenvironment than in the periphery. All of our tumor microenvironment activators, as we call them, seek to more effectively reactivate these tumor-reactive T cells than existing therapies. This design is meant to drive improved tolerability at higher doses compared to the dosing of separate anti-CTLA-4 and anti PD-1 antibodies, for example, which has delivered better responses at the cost of tolerability.

In our first 6 dose escalation cohorts, we observed that XmAb20717 to be generally well tolerated in heavily pretreated patients. Consistent with our hypothesis of inhibiting both PD-1 and CTLA-4, we observed robust dose-dependent increases in biomarkers of T cell activation and pharmacodynamic activity consistent with blockade of both receptors. It was also encouraging to observe cases of clinical activity as we moved into the higher doses cohorts, which we detailed in the press release in May. Based on these data, we opened expansion cohorts in several tumor types at 10 milligrams per kilogram as well as additional dose escalation cohorts starting at 15 milligrams per kilogram as we did not reach the maximum tolerated dose. Expansion cohorts for patients with melanoma and advanced non-small cell lung cancer are fully enrolled. We look forward to sharing continued progress from the 20717 program as well as our other tumor microenvironment targeting bispecific antibody programs in Phase I studies.

XmAb2314 targets PD-1 and the co-stimulatory receptor, ICOS, and XmAb22841, which targets the checkpoint CTLA-4 and LAG-3, the latter which has begun dosing patients in combination with pembrolizumab.

Moving on to our clinical stage T cell engagers. These are tumor-targeted bispecific antibodies that contain both the tumor antigen binding domain and the cytotoxic T cell binding domain, specifically CD3 binding domain. These CD3 bispecifics activate T cells at the site of the tumor in order to potentially kill malignant cells. We continue to dose patients in our Phase I studies of vibecotamab, which targets CD123 and acute myeloid leukemia and plamotamab which targets CD20 in B-cell malignancies. And as we have previously disclosed, we plan to initiate additional clinical programs, subject to impacts from the COVID-19 pandemic, likely next year.

We also continue to dose patients in the Phase I study of tidutamab, which targets the somatostatin receptor 2, and we expect that we will present initial data from these ongoing -- this ongoing study in patients with neuroendocrine tumors in the second half of this year.

Finally, we're developing a suite of cytokines, which are immune signaling protein that are built on the XmAb bispecific Fc domain and incorporate the Xtend technology. Using our Fc domain and tuning the potencies enabled cytokines with improved drug like properties, such as slower receptor mediated clearance and longer half-life and potentially superior tolerability. Our first cytokine program and lead in our collaboration with Genentech is XmAb24306, which they call RG6323. It's an IL-15/IL-15 receptor alpha complex fused with our bispecific Fc domain. It targets the expansion and activation of T cells and natural killer cells.

Genentech is currently enrolling patients in a Phase I study evaluating XmAb24306 and quickly moving in combination with atezolizumab their anti-PD-L1 antibody. We plan to explore a number of our own combination studies pending completion of the initial dose escalation study. We also look forward to keeping you informed about all our clinical programs as they progress.

Now I'll hand the call over to John Kuch who will review the second quarter and first 6 months financial results. John?

--------------------------------------------------------------------------------

John J. Kuch, Xencor, Inc. - Senior VP & CFO [6]

--------------------------------------------------------------------------------

Thank you, Allen.

Xencor continues to maintain its strong financial position, which enables us to support our portfolio of clinical research stage bispecific antibody and cytokine drug programs. Our diversified portfolio of partnerships and collaborations continue to provide us with upfront payments, milestones and royalties, important sources of nondiluted capital. With the FDA approval of MorphoSys Monjuvi last Friday, we will receive a $25 million milestone payment which we will recognize as revenue in the third quarter. As a reminder, we are also eligible to receive royalties on worldwide net sales in the high single to low double-digit percent range and additional development, regulatory and sales milestone payments.

At June 30, 2020, our cash, cash equivalents, marketable and equity securities totaled $587.4 million compared to $601.3 million at December 31, 2019. The decrease reflects cash used to fund operating activities in the first 6 months of 2020, offset by upfront payments, milestone payments and royalties from our partnership and licensing arrangements.

For the second quarter of 2020, revenues were $13.1 million compared to $19.5 million for the same period in 2019. These revenues include royalty revenue from Alexion and licensing revenue from Gilead compared to the same period in 2019

where revenues primarily reflect research collaboration revenue from Genentech and Astellas and milestone revenue from Alexion.

For the first 6 months of 2020, revenues were $45.5 million compared to $131.4 million for the same period in 2019. Our revenues in 2020 include royalty revenue from Alexion, milestone revenue from MorphoSys and licensing revenue from our Gilead and Aimmune collaborations compared to licensing and collaboration revenue earned from Genentech and Astellas in 2019.

Research and development expenditures for the second quarter of 2020 were $43.5 million compared to $33.3 million for the same period in 2019. After the first 6 months in 2020, they were $77.4 million compared to $61.5 million for the same period in 2019. The increases in R&D is primarily due to increased spending on our plamotamab and XmAb20717 clinical programs as well as our preclinical IL-2 cytokine program, XmAb27564 and our preclinical ENPP3 x CD3 2+1 bispecific antibody program, XmAb30819, both of which we have advanced into IND-enabling activities. We note that there was lower spending in 2020 on our XmAb24306 and obexelimab programs.

General and administrative expenses for the second quarter of 2020 were $7.2 million compared to $5.8 million in the same period in 2019. For the first 6 months in 2020, G&A expenses were $14.4 million compared $11.3 million for the same period in 2019. Additional spending here is primarily due to increased staffing and spending on professional fees.

The net loss for the second quarter of 2020 was $35 million or $0.61 on a fully diluted per share basis compared to a net loss of $16 million or $0.28 on a fully diluted per share basis for the same period in 2019. The higher net loss reported in 2020 is primarily due to lower partnership and collaboration revenue and higher R&D expenses in 2020.

For the first 6 months in 2020, net loss was $43.1 million or $0.76 on a fully diluted per share basis compared to net income of $64 million or $1.10 on a fully diluted per share basis for the same period in 2019.

Our net loss for the first 6 months of 2020 compared to the net income reported for same period in 2019 is primarily due to revenue recognition from our Genentech collaboration 2019.

Noncash stock-based compensation expense for the first 6 months of 2020 was $14.7 million compared to $15.2 million for the same period in 2019. Total shares outstanding were 57.2 million as of June 30, 2020, compared to 56.5 million as of June 30, 2019.

Based on current operating plans, Xencor expects to have cash to fund research and development programs and operations into 2024. Xencor expects to end 2020 with between $525 million and $575 million in cash, cash equivalents, marketable securities and equity securities.

With that, we'd now like to open up the call for your questions. Operator?

================================================================================

Questions and Answers

--------------------------------------------------------------------------------

Operator [1]

--------------------------------------------------------------------------------

(Operator Instructions)

Our first question is from Ted Tenthoff with Piper Sandler.

--------------------------------------------------------------------------------

Edward Andrew Tenthoff, Piper Sandler & Co., Research Division - MD & Senior Research Analyst [2]

--------------------------------------------------------------------------------

So congratulations on the tafasitamab approval. I'm wondering -- give us a sense of what the royalties are and whether there are other future milestones beyond the approval milestone for other indications and things like that.

--------------------------------------------------------------------------------

Bassil I. Dahiyat, Xencor, Inc. - Co-Founder, CEO, President & Director [3]

--------------------------------------------------------------------------------

Sure. Thanks. And I hope you're staying safe, Ted, out with that tropical storm in New York along with all the other new Yorkers.

So the royalties are high single to low double digit, and they're tiered. That's the most detail we're allowed to share at this point. They're worldwide royalty, so you can see they're worldwide sales, regardless of whether the company selling is Incyte or MorphoSys and, of course, Incyte is ex U.S. commercial rights. There are significant milestones for both developments in other indications within oncology as well as non oncology, though there's no development going on for that at the moment that we're aware of. So there's other oncology indication, regulatory -- development regulatory milestones and there are sales milestones.

John, do you want to give a little bit of granularity on the magnitude of those?

--------------------------------------------------------------------------------

John J. Kuch, Xencor, Inc. - Senior VP & CFO [4]

--------------------------------------------------------------------------------

Yes. The sales milestones are $50 million, and the other development, regulatory are anywhere in the $50 million, $75 million range.

--------------------------------------------------------------------------------

Bassil I. Dahiyat, Xencor, Inc. - Co-Founder, CEO, President & Director [5]

--------------------------------------------------------------------------------

Yes, depending on which ones we...

--------------------------------------------------------------------------------

John J. Kuch, Xencor, Inc. - Senior VP & CFO [6]

--------------------------------------------------------------------------------

Read more:
Edited Transcript of XNCR.OQ earnings conference call or presentation 4-Aug-20 8:30pm GMT - Yahoo Finance

Humanigen to Present at the BTIG Virtual Biotechnology Conference 2020 – Business Wire

BURLINGAME, Calif.--(BUSINESS WIRE)--Humanigen, Inc., (HGEN) (Humanigen), announced today that Cameron Durrant, MD, MBA, Chief Executive Officer and Dale Chappell, MD, MBA, Chief Scientific Officer of Humanigen will present a company overview and business update at the BTIG Virtual Biotechnology Conference 2020 on Tuesday, August 11th, 2020 at 2:00 pm Eastern Time. The conference is being held in a virtual format.

A live webcast of the event can be accessed at https://www.humanigen.com/investor-materials.

An archived replay of the event will be available on the Company website for 30 days following the event.

About Humanigen, Inc.

Humanigen, Inc. is developing its portfolio of clinical and pre-clinical therapies for the treatment of cancers and infectious diseases via its novel, cutting-edge GM-CSF neutralization and gene-knockout platforms. We believe that our GM-CSF neutralization and gene-editing platform technologies have the potential to reduce the inflammatory cascade associated with coronavirus infection. The companys immediate focus is to prevent or minimize the cytokine release syndrome that precedes severe lung dysfunction and ARDS in serious cases of SARS-CoV-2 infection. The company is also focused on creating next-generation combinatory gene-edited CAR-T therapies using strategies to improve efficacy while employing GM-CSF gene knockout technologies to control toxicity. In addition, the company is developing its own portfolio of proprietary first-in-class EphA3-CAR-T for various solid cancers and EMR1-CAR-T for various eosinophilic disorders. The company is also exploring the effectiveness of its GM-CSF neutralization technologies (either through the use of lenzilumab as a neutralizing antibody or through GM-CSF gene knockout) in combination with other CAR-T, bispecific or natural killer (NK) T cell engaging immunotherapy treatments to break the efficacy/toxicity linkage, including to prevent and/or treat graft-versus-host disease (GvHD) in patients undergoing allogeneic hematopoietic stem cell transplantation (HSCT). Additionally, Humanigen and Kite, a Gilead Company, are evaluating lenzilumab in combination with Yescarta (axicabtagene ciloleucel) in patients with relapsed or refractory large B-cell lymphoma in a clinical collaboration. For more information, visit http://www.humanigen.com.

See the rest here:
Humanigen to Present at the BTIG Virtual Biotechnology Conference 2020 - Business Wire

StemCells 21 Premium Stem Cell Treatments In Bangkok …

We organize our stem cell packages based on the size and complexity of your condition. Small, simple conditions require relatively few stem cells to treat them. Large, complicated conditions or conditions you wish to treat as quickly and as completely as possiblewill require larger volumes of stem cells to get the job done.

We currently offer four stem cell packages.

Condition Level

Treatment Type

Stem Cell Volume

I

Single Area of Focus & Treatment

50,000,000 stem cells

II

Multiple Areas of Focus, Simple Inflammatory Conditions, or Early Stage Disease

100,000,000 stem cells

III

Complex Conditions or Premium Option for Simple Conditions

200,000,000 stem cells

IV

Advanced Stage Disease or Fastest Possible Treatment for All Conditions

300,000,000+ stem cells

We select your supportive therapies based on your condition. Your supportive therapies will direct your stem cells to the impacted tissue that needs to be regenerated, trigger selective cell activity, and provide your body with additional biological building blocks to deliver a faster and more complete regeneration.

We offer many supportive therapies, that include but are not limited to:

We curate an additional set of simple supportive therapies that you will bring home with you after your treatment, and self-administer for one to three months. This take home set ensures that your stem cells and your impacted tissue continue to receive direction and building blocks to continue to regenerate after your initial treatment.

We typically provide a one to three-month supply of:

Read the original post:
StemCells 21 Premium Stem Cell Treatments In Bangkok ...

Newly discovered epigenetic tag offers insight into …

Scientists from the Babraham Institute have gained a new understanding of how molecular signals and switches control how an embryo develops into an adult. The new research, published today in the journal Nature, details how a newly discovered form of epigenetic regulation controls the development of embryonic stem (ES) cells.

The research, funded by BBSRC, MRC, the University of Cambridge and EPIGENOME, has important implications for regenerative medicine as it could offer new methods for controlling how ES cells differentiate in every cell in the human body and, potentially, to the growing field of induced pluripotent stem (iPS) cells where adult stem cell are reprogrammed. Embryonic stem (ES) cells are pluripotent cells present in the early embryo, which have the capacity to differentiate into all the specialised cells that make up the adult body. As an embryo develops, the cells respond to signals and differentiate to acquire a particular fate, for example a skin cell.

Cell fate is governed not only by the genome, but also by chemical changes to DNA that alter the DNA structure but not its sequence. These epigenetic tags are one of the ways that genes get switched on or off in different places at different times, enabling different tissues and organs to arise from a single fertilised egg and also helps to explain how our genes can be influenced by the environment.. The new research reveals that a new type of epigenetic modification, 5-hydroxymethylcytosine (5hmC), plays a critical role mediating the external signals that instruct a cell how to develop; this tiny chemical tag (5hmC) is attached to or removed from the genetic sequence depending on the message received, switching genes on or off.

The researchers managed to identify the location of this tag throughout the genome, using high throughput sequencing methods. They observed for so called pluripotency-related genes that, as 5hmC decreases, another previously known epigenetic modification, 5-methylcytosine (5mC) increases this shift has consequences in determining how genes function and hence a cells developmental fate. The pluripotency window for stem cells is short-lived but essential for the environment and pre-defined genetic programme to exert influence on the direction that each cell should take to build a healthy embryo.

Hydroxy-methylation appears to be linked to a higher degree of pluripotency; when the process of generating 5hmC tags in the stem cell genome was disrupted, the researchers saw the pluripotencyrelated genes were down-regulated, causing the cells to be more receptive to signals that promote differentiation than would normally be the case for stem cells. The two epigenetic modifications, 5mC and 5hmC, were seen to have other opposing behaviour in the genome, which might be important for maintaining flexibility of stem cells in order to respond accurately to external cues.

Knowing how hydroxymethylation works in embryonic stem cells might also help with reprogramming adult cells into induced pluripotent stem cells (iPS cells), since removal of methylation is important in generating these cells. Hence increasing the amounts of hydroxymethylation during reprogramming might make the process more efficient and error-free. This might help with developing improved strategies for regenerative medicine.

Professor Wolf Reik, who led the study at the Babraham Institute, which receives strategic funding from the Biotechnology and Biological Sciences Research Council (BBSRC) said, This work provides an exciting new perspective on what makes embryonic stem cells special. It shows how the balance between opposing epigenetic marks is important for the ability of stem cells to differentiate into different tissues. We may be able to use the new epigenetic mark, hydroxymethylation, for improved strategies for reprogramming any cell into a stem cell, and hence in regenerative medicine.

While advancing our understanding of the biology behind reprogramming, these findings may also help to explain how epigenetic changes occurring during ageing can cause disease, since conditions like heart disease and autoimmune disorders may be associated with failure of epigenetic regulation. It is known that 5hmC is most abundant in ES cells and in the brain. This study opens up many questions on the role that 5hmC may play in a non-dividing brain cell, modulating gene expression, and its relationship with memory formation and neurological disorders.

Gabriella Ficz, joint lead author of this research said, Our work reveals important aspects about the epigenetics of stem cells but looking at our data I couldnt stop wondering about the involvement of this new modification in ageing and complex diseases like diabetes, autoimmune disorders and schizophrenia as well as cancer and obesity. It is an exciting time for epigenetic research!

Miguel R. Branco, joint lead author commented, The recent discovery of this new DNA modification has attracted a quickly growing interest from the scientific community. Whilst it is still early days and we will have to dig deeper to better understand its role, our work has unveiled important links between hydroxymethylation, methylation and the regulation of pluripotency genes. Professor Douglas Kell, BBSRC Chief Executive, said, Fundamental biological processes such as epigenetic regulation have important and far-reaching consequences. As this research shows, epigenetics offers both the potential to underpin new therapies in the future but also to help us to understand how the normal functioning of our bodies operates.

The Babraham Institute undertakes world-leading life sciences research to generate new knowledge of biological mechanisms underpinning ageing, development and the maintenance of health.Professor Michael Wakelam, Director of the Babraham Institute, said, These innovative studies from the Reik laboratory are part of the Babraham Institutes central mission to understand lifelong health and wellbeing. This research at Babraham was supported by the BBSRC, the MRC, the University of Cambridge and by the EPIGENOME Network of Excellence.

ublication details: Ficz G, Branco MR, Seisenberger S, Santos F, Krueger F, Hore TA, Marques CJ, Andrews SR, Reik W (In press) Dynamic regulation of 5-hydroxymethylcytosine in mouse ES cells and during differentiation. Nature http://dx.doi.org/10.1038/nature10008

Contact details: The Knowledge Exchange Office Email:kec@babraham.ac.uk Tel: +44 (0)1223 496206

The Babraham Institute Babraham Research Campus Cambridge CB22 3AT United Kingdom Notes to Editors: About the Babraham Institute: The Babraham Institute undertakes world-class life sciences research to generate new knowledge of biological mechanisms underpinning ageing, development and the maintenance of health. Our research focuses on cellular signalling, gene regulation and the impact of epigenetic regulation at different stages of life. By determining how the body reacts to dietary and environmental stimuli and manages microbial and viral interactions, we aim to improve wellbeing and support healthier ageing. The Institute is strategically funded by the Biotechnology and Biological Sciences Research Council (BBSRC), part of UK Research and Innovation, through an Institute Core Capability Grant and also receives funding from other UK research councils, charitable foundations, the EU and medical charities. Website: http://www.babraham.ac.uk The Biotechnology and Biological Sciences Research Council(BBSRC)is the UK funding agency for research in the life sciences. Sponsored by Government, BBSRC annually invests around 450 million in a wide range of research that makes a significant contribution to the quality of life for UK citizens and supports a number of important industrial stakeholders including the agriculture, food, chemical, health and well-being and pharmaceutical sectors. BBSRC carries out its mission by funding internationally competitive research, providing training in the biosciences, fostering opportunities for knowledge transfer and innovation and promoting interaction with the public and other stakeholders on issues of scientific interest in universities, centres and institutes. Website: bbsrc.ukri.org/ Babraham Bioscience Technologies Ltdis responsible for managing the Babraham Research Campus Bioincubator. BBT brings together all the elements to support innovation and enable the successful exploitation of research in the biomedical sector based on technologies emanating from the Babraham Institute and bioventures relocating to the campus. BBT has taken a prominent role regionally, initiating and leading partnerships to promote knowledge and skills flow and has established a reputation for successfully translating innovative science into viable business opportunities through partnerships for wealth creation.

Website:www.babraham.com

Read more from the original source:
Newly discovered epigenetic tag offers insight into ...

NICE Amends Guidance on Haematopoietic Stem Cell Transplantation During the Pandemic – Medscape

The National Institute for Health and Care Excellence (NICE) has amended its recommendations on advice and testing for COVID-19 among patients undergoing haematopoietic stem cell transplantation and donors.

As of 29 July 2020,changeshave been made to the following sections:

Advice for patients to limit the number of family members who attend appointments (recommendation 1.3) and explaining measures to limit infection risk (new recommendation 1.4).

Advice for patients on minimising risk of respiratory infections before transplantation (recommendation 3.1).

Testing for respiratory viruses before transplantation (recommendation 3.2).

Additional investigations for patients who test positive for or are suspected of having COVID-19 (new recommendation 3.7).

Tests for donors and actions if the results are positive (new recommendation 4.5 and recommendation 4.6); these recommendations now apply to related donors, not just sibling donors (recommendation 4.1).

Risk assessment for donors who test positive (recommendation 4.8) and a reduction in the delay in providing blood products after a positive test (recommendation 4.10).

Advice for patients post-transplant (recommendation 5.2).

Assessing when staff who test positive or have symptoms can return to work (recommendation 6.2).

Routine screening for staff (new recommendation 6.3).

Prioritising treatment (table 1).

Risk assessments for ambulatory transplant pathways (new recommendation 8.3).

What to do when a centre is temproarily closed (recommendation 8.6).

Assessing risk in storing cells from a donor with COVID-19 (recommendation 8.9) and the viability of cryopreserved stem cells (new recommendation 8.10).

Using granulocyte-colony stimulating factor to minimise the use of chemotherapy priming.

NICE has also removed recommendations (originally numbered 3.3, 3.4 and 7.3) that advised deferring most autologous and allogeneic haematopoietic stem cell transplants, and deferring transplants if further treatment or immunosuppression would put them at more risk from COVID-19 in the community. This is to reflect changes in the risk of infection and the capacity in services.

This article originally appeared on Univadis, part of the Medscape Professional Network.

Read the original:
NICE Amends Guidance on Haematopoietic Stem Cell Transplantation During the Pandemic - Medscape

Cure for Covid-19 may never materialise says WHO, but there is still hope – The National

In March, when Covid-19 was declared a pandemic by the World Health Organisation, few people imagined that the outbreak would stretch into the summer. Many political leaders were hopeful that temporary lockdowns across the world would end the crisis and that life could soon go back to normal. Yet, in the absence of a vaccine, precautions such as wearing masks, working from home and physical distancing have become the new norm.

On Monday, WHO director general Tedros Adhanom Ghebreyesus warned that a solution may, in fact, never be found. When it comes to the coronavirus, he said: there is no silver bullet at the moment and there might never be, despite the worlds best efforts to find a cure or a treatment for Covid-19.

It is the WHOs responsibility to inject a dose of reality into global health policies and to manage expectations. Vaccines can take years to develop and test until they are safe for the public to use, but this does not mean that the coronavirus crisis cannot be controlled until that happens.

Research into treatments and cures are already well under way. Across the world there are 23 vaccines being tested on humans, but only three of them have entered the last phase. The UAE, having understood the importance of research into the coronavirus early on, invested in it heavily. It is the first country in the world that began phase III clinical trials and thousands of people volunteered to take the jab here in Abu Dhabi.

While the public awaits a vaccine, there is hopeful news: treatments to alleviate the suffering of patients and hasten their recovery have shown promising results.

In May, Abu Dhabi Stem Cell Centre said it had developed a new type of stem cell therapy that could help shorten recovery time for Covid-19 patients. This treatment is free for those suffering from the disease in the UAE. In the US, meanwhile, the antiviral drug Remdesivir has managed to improve the health of patients who are severely affected by Covid-19.

It is the WHOs responsibility to inject a dose of reality into global health policies and manage expectations

While scientists are at work to find a vaccine or a treatment, the knowledge they have gathered so far about Covid-19 has equipped governments and the public to fight the pandemic more efficiently. Directives on wearing masks, physical distancing and actively sanitising are among measures now understood and adopted widely. Lockdowns, despite their negative economic impact, have slowed the spread of the virus, especially when cases peaked.

As nations gradually reopen, governments are more aware of the tools needed to prevent a public health catastrophe. Mass testing and equipping hospitals with life-saving personal protective equipment and ventilators are a must. In the UAE, enforcing such measures has allowed the country to attain a 90 per cent coronavirus cure rate, as 5 million tests have been carried out nationwide.

While the coronavirus is unlikely to be eradicated in the near future, the world has learnt to better control outbreaks. A time will hopefully come when the pandemic is behind us. Till then, however, we must use and perfect the tools and knowledge gathered in the past eight months to continue fighting the coronavirus.

Updated: August 4, 2020 09:03 PM

Read the original here:
Cure for Covid-19 may never materialise says WHO, but there is still hope - The National

Teams Working to Image Protein Clumps in Parkinson’s Share $8.5M Prize – Parkinson’s News Today

The Michael J. Fox Foundation (MJFF) has awarded a total of $8.5 million to research teams at AC Immune, Mass General Brigham, and Merck to develop a way to detect the protein alpha-synuclein in the brains of people living with Parkinsons disease.

The three are winners of theKen Griffin Alpha-synuclein Imaging Competition, which was launched in September 2019 to promote research into a potential diagnostic tool for Parkinsons.

Alpha-synuclein is believed to play a role in the onset of Parkinsons symptoms. The protein forms abnormal aggregates, called Lewy bodies,in the brains of people with the disease, as well as in those brains of those with Lewy body dementia.

Through current methods, it is only possible to detect the presence of alpha-synuclein and Lewy bodies in brain tissue collected post-mortem.MJFF has supported research into its detection in living patients for over a decade.

The competitions goal is to find what is called a tracer a molecule that allows for the identification of a specific target.

In this instance, the tracer would be for alpha-synuclein and included in a positron emission tomography (PET) scan, which already uses a radioactive tracer to generate a 3D image of the brain.

Most of the prize money, $7.5 million, was donated by Ken Griffin, the founder and CEO of the investment firm Citadel.

I am proud to join The Michael J. Fox Foundation in supporting the important research driven by these incredible teams at AC Immune, Mass General Brigham and Merck, Griffin said in a press release. We look forward to their continued progress as we work together to unlock game-changing breakthroughs for the millions of people living with Parkinsons disease.

Using its proprietary Morphomer discovery platform, researchers at AC Immune have isolated two potential small molecules for alpha-synuclein detection and moved them into clinical trials.AC Immune intends to use the grant money to continue its preclinical and clinical program to optimize and test an alpha-synuclein tracer compound, working with researchers at Lund University and Skne University Hospital.

Researchers at Mass General Brigham plan to use the funding for a genetic approach toward alpha-synuclein detection.The team will run a high-throughput (automated to maximize efficiency) screen of a library of DNA, allowing for the rapid evaluation of billions of tracer candidates.

Candidate molecules identified will then be investigated using post-mortem tissue and cell cultures derived from patients stem cells.

Researchers at Merck (known as MSD outside the U.S. and Canada) have already identified several potential small molecules that could be used as alpha-synuclein tracers, and intend to use the new funding to optimize such potential tracers.

Once optimization is complete, the Merck team intends to launch clinical trials of the most promising tracer candidates.

Under this competition, the team that makes the most progress over the next two years will be awarded an additional $1.5 million to support its work.

David earned a PhD in Biological Sciences from Columbia University in New York, NY, where he studied how Drosophila ovarian adult stem cells respond to cell signaling pathway manipulations. This work helped to redefine the organizational principles underlying adult stem cell growth models. He is currently a Science Writer, as part of the BioNews Services writing team.

Total Posts: 208

Ana holds a PhD in Immunology from the University of Lisbon and worked as a postdoctoral researcher at Instituto de Medicina Molecular (iMM) in Lisbon, Portugal. She graduated with a BSc in Genetics from the University of Newcastle and received a Masters in Biomolecular Archaeology from the University of Manchester, England. After leaving the lab to pursue a career in Science Communication, she served as the Director of Science Communication at iMM.

Here is the original post:
Teams Working to Image Protein Clumps in Parkinson's Share $8.5M Prize - Parkinson's News Today

Nerve Repair and Regeneration Market worth $9.7 billion by 2025 – Exclusive Report by MarketsandMarkets – PRNewswire

CHICAGO, Aug. 4, 2020 /PRNewswire/ -- According to the new market research report "Nerve Repair and Regeneration Marketby Products (Nerve Conduits, Nerve Wraps, Vagus Nerve Stimulation, Sacral Nerve Stimulation, Spinal Cord Stimulation, TENS, TMS), Application (Neurorrhaphy, Nerve Grafting, Stem Cell Therapy) and Region - Global Forecast to 2025", published by MarketsandMarkets,the global Nerve Repair and Regeneration Marketsize is projected to reach USD 9.7 billion by 2025 from USD 6.3 billion in 2020, growing at a CAGR of 9.1% from 2020 to 2025.

Download PDF Brochure: https://www.marketsandmarkets.com/pdfdownloadNew.asp?id=883

The Growth in Nerve Regeneration Marketis driven mainly by high incidence of nerve injuries, the growing prevalence of neurological disorders, and rising government support for neurologic disorder research.

Neurostimulation and Neuromodulation Devices accounted for the largest share of the market, by product, in 2019

By product, the nerve repair market is segmented into neurostimulation and neuromodulation devices and biomaterials. The neurostimulation and neuromodulation devices segment is segment is expected to grow at the highest growth rate during the forecast period. The large market share of this segment is driven mainly by rising government expenditure for neurologic disorders, and favorable reimbursement .

Browsein-depth TOC on"Nerve Repair and Regeneration Market"

142 Tables 43 Figures 167 Pages

By neurostimulation and neuromodulation application, internal neurostimulation and neuromodulation accounted for the largest market share in 2019

Based on the neurostimulation and neuromodulation application , the Nerve Regeneration Market is segmented the neurostimulation and neuromodulation devices market is segmented into internal neurostimulation and neuromodulation applications and external neurostimulation and neuromodulation applications. The internal neurostimulation and neuromodulation segment is estimated to register the highest CAGR during the forecast period. This can primarily be attributed to the increasing incidence of neurological disorders across the globe.

By Biomaterials application, direct nerve repair/neurorrhaphy accounted for the largest market share in 2019

Based on application, the biomaterials market is segmented into direct nerve repair/neurorrhaphy, nerve grafting, and stem cell therapy. In 2019, the direct nerve repair segment accounted for the largest share of the market. This can be attributed to the increasing incidence of neurological disorders across the globe.

Get 10% Customization on this Research Report: https://www.marketsandmarkets.com/requestCustomizationNew.asp?id=883

North America was the largest regional market for Nerve Repair Market in 2019

The Nerve Repair And Regeneration Market is segmented into five major regions, namely, Europe, North America, the Asia Pacific, Latin America, and Middle East & Africa. In 2019, North America accounted for the largest share of the Nerve Regeneration Market, followed by Europe. The rising incidence of neurological disorders, favorable reimbursement policies, and the strong presence of industry players in the region are the major factors driving the growth of the market in North America.

Some of the major players operating in the global Nerve Repair And Regeneration Market include Medtronic, PLC. (Ireland), Boston Scientific Corporation (US), Abbott Laboratories (US), AxoGen, Inc. (US), Baxter International, Inc. (US), LivaNova, PLC. (UK), Integra LifeSciences (US), Polyganics (Netherlands), NeuroPace, Inc. (US), Soterix Medical, Inc. (US), Nevro Corp (US), Synapse Biomedical, Inc. (US), Aleva Neurotherapeutics (Switzerland), Collagen Matrix, Inc. (US), KeriMedical (Switzerland), BioWave Corporation (US), NeuroSigma (US), tVNS Technologies GmbH(Germany), and GiMer Medical (Taiwan).

Browse Adjacent Markets:Medical Devices Market Research Reports & Consulting

Get Special Pricing on Bundle Reports: https://www.marketsandmarkets.com/RequestBundleReport.asp?id=883

Browse Related Reports:

Neuromodulation Marketby Technology- Internal (Deep Brain Stimulation, Vagus Nerve Stimulation), External (Transcranial Magnetic Stimulation), Application (Ischemia, Chronic Pain, Parkinson's, Depression, Tremor, Epilepsy, Migraine) - Global Forecast to 2025

https://www.marketsandmarkets.com/Market-Reports/neurostimulation-devices-market-921.html

Neurodiagnostics Marketby Product (Diagnostic & Imaging Systems (MRI, Ultrasound), Clinical Testing (PCR, NGS), Reagents & Consumables), Disease Pathology (Epilepsy, Stroke), End User, and Region - Global Forecast to 2024

https://www.marketsandmarkets.com/Market-Reports/neurodiagnostics-market-229090425.html

About MarketsandMarkets

MarketsandMarkets provides quantified B2B research on 30,000 high growth niche opportunities/threats which will impact 70% to 80% of worldwide companies' revenues. Currently servicing 7500 customers worldwide including 80% of global Fortune 1000 companies as clients. Almost 75,000 top officers across eight industries worldwide approach MarketsandMarkets for their painpoints around revenues decisions.

Our 850 fulltime analyst and SMEs at MarketsandMarkets are tracking global high growth markets following the "Growth Engagement Model GEM". The GEM aims at proactive collaboration with the clients to identify new opportunities, identify most important customers, write "Attack, avoid and defend" strategies, identify sources of incremental revenues for both the company and its competitors. MarketsandMarkets now coming up with 1,500 MicroQuadrants (Positioning top players across leaders, emerging companies, innovators, strategic players) annually in high growth emerging segments. MarketsandMarkets is determined to benefit more than 10,000 companies this year for their revenue planning and help them take their innovations/disruptions early to the market by providing them research ahead of the curve.

MarketsandMarkets's flagship competitive intelligence and market research platform, "Knowledge Store" connects over 200,000 markets and entire value chains for deeper understanding of the unmet insights along with market sizing and forecasts of niche markets.

Contact:

Mr. Aashish Mehra MarketsandMarkets INC. 630 Dundee Road Suite 430 Northbrook, IL 60062 USA: +1-888-600-6441 Email: [emailprotected] Research Insight: https://www.marketsandmarkets.com/ResearchInsight/nerve-repair-regeneration-market.asp Visit Our Website: https://www.marketsandmarkets.com Content Source: https://www.marketsandmarkets.com/PressReleases/nerve-repair-regeneration.asp

SOURCE MarketsandMarkets

Originally posted here:
Nerve Repair and Regeneration Market worth $9.7 billion by 2025 - Exclusive Report by MarketsandMarkets - PRNewswire

Induced Pluripotent Stem Cells: The Future of Tissue …

As viable human brain tissue is not available for use in studying disease development and creating therapies for neurological disorders like Huntingtons disease (HD), researchers desperately needed an alternative cell source for this purpose. Embryonic stem cells fit this role but have many disadvantages, especially for treatments, including immune rejection by the recipient. Some of these drawbacks have been overcome by a recent discovery that revolutionized the face of stem cell biology. In 2006, Shinya Yamanakas research group at Kyoto University made a groundbreaking announcement: they had discovered that adult cells could be genetically engineered to revert back to apluripotent, stem cell-like state. As iPSC (induced pluripotent stem cell) production rapidly improved, the cells were soon able to compete with traditional fetal, embryonic, and adult stem cells. The primary advantages of iPSCs compared to other stem cells are: a) iPSCs can be created from the tissue of the same patient that will receive the transplantation, thus avoiding immune rejection, and b) the lack of ethical implications because cells are harvested from a willing adult without harming them. These patient-specific cells can be used to study diseases in vitro, to test drugs on a human model without endangering anyone, and to hopefully act as tissue replacement for diseased and damaged cells.

Like other stem cells, iPSCs have the ability to proliferate indefinitely in vitro, creating a theoretically unlimited source of cells. Like embryonic stem cells, iPSCs can also differentiate into any cell of the body, regardless of the original tissue from which they are created. Scientists have found how to direct the differentiation of pluripotent stem cells into many types of target tissue, including neural tissue. iPSCs demonstrate that by the introduction of just four genes into somatic cells that normally cannot differentiate at all, cells can be created that can differentiate into every cell type in the body. The early results of iPSC differentiation studies look promising. For example, human fibroblasts have been successfully turned into iPSCs that then are differentiated into insulin-producing cells, a result that holds much potential for the treatment of diabetes. Mouse iPSCs have been differentiated into cardiovascular (heart muscle) cells, that actually show the contractile beating expected of heart tissue.

Although there are many problems that still must be addressed for iPS technology, such as the tendency for tumors to evolve after iPSC transplantation and the low efficiency of the technology, iPSCs could completely change how diseases are approached in biomedical research. For HD and other neurological disorders, iPSCs could create perfect models for the cells of the central nervous system that are harmed in the diseases.

Stem cell biology is a very hot topic in modern medicine, yet much is still unknown about the mechanisms underlying pluripotency and differentiation. In order for safe, controllable, and efficient cellular reprogramming to be achieved, there must be more knowledge on the regulation of stem cell states and transitions. iPSCs show that specialized cells and tissue can be transformed into other types of cells, proving cells are much more flexible than previously thought. As the study of HD will greatly benefit from this new, unlimited source of neural cells for research and cell therapy, iPSCs may be able to provide new and innovative treatments for HD.

The creation of pluripotent cells has been widely studied for decades. In 1976, the first method of fusion of an adult somatic cell with embryonic cells to create pluripotent stem cells was reported. However, fusion with embryonic cells created unstable cells that were rejected by immune systems after transplantation. If the genes that induced pluripotency could be isolated from their parent embryos and injected into somatic cells, these problems could be avoided.

Yamanakas research team studied twenty-four genes expressed by embryonic stem cells in an effort to track down these essential genes that induce pluripotency. To detect pluripotency, they looked for cells expressing genes that were traditionally expressed only in embryos. They discovered that the addition of four genes induced a cell into a pluripotent state capable of then becoming many different cell types.

Subsequent studies showed that other gene combinations were also successful in reengineering cells into iPSCs, but none were as efficient as the first four. Adding other genes that are expressed in early development was shown to increase reprogramming efficiency, and the specific genes needed varied depending on the cell type that was being forced back to its pluripotent state. As the four factors and their alternatives were largely discovered by trial and error, it is not known how the genes induce pluripotency. Discovering how genes work may point to ways of improving the efficiency of the process and assessing the quality of iPSCs.

The specific genes that induce iPSCs tell scientists a lot about the characteristics of the cells themselves. Pluripotent stem cells are very closely related to tumor cells. Both can survive and proliferate indefinitely, and a test of pluripotency is whether a cell can create a tumor. It is therefore no surprise that two tumor-related genes, c-Myc and Klf4, are needed to create iPSCs. Another requirement of pluripotent stem cells is open and active chromatin structures (for more information on chromosomes, click here and DNA transcription click here). The c-Myc gene codes for proteins that loosen the chromatic structure, stimulating differentiation. Klf4 impedes proliferation. c-Myc and Klf4 in this way regulate the balance between proliferation and differentiation. If only c-Myc and Klf4 are used in the engineering of iPSCs, tumor cells will ariseinstead of pluripotent stem cells. Oct3/4 and Sox2 are required to direct cell fate towards a more embryonic stem cells (ESC)-like phenotype. Oct3/4 directs specific differentiation, such as neural and cardiac differentiation, while Sox2 maintains pluripotency. Oct3/4 and Sox2 together ensure that iPSCs are indeed pluripotent stem cells and not tumor cells.

The programming of iPSCs depends both on the original cell type being transformed and the levels of each reprogramming factor that is expressed. Expressing Oct3/4 more than the other genes increases efficiency. Increasing the expression of any of the other three genes decreases the efficiency. There is clearly a correlation between gene expression ratio and reprogramming efficiency, but the optimal ratio is likely to vary depending on the cell type being reprogrammed. For instance, when neural progrenitor cells are reprogrammed, they do not require Sox2 as they express this gene sufficiently already. The level of expression of other important genes for maintaining pluripotency also can affect the reprogramming process and the quality of the resulting cells.

The effect gene expression ratio has on reprogramming may explain why efficiency is typically so low (less than 1% of cells are reprogrammed successfully). Reprogramming is a slow process, and so the timing of various events may also exert a great influence over thecells success. The minimum time for the full reprogramming of a mouse somatic cell into an iPSC is between eight and twelve days. The timing of the mechanism for cellular reprogramming may also be a reason for low efficiency, as the cells can only proceed if the right molecular events happen in the correct order.

In the first studies of iPSCs, the cells were shown to be similar to ESCs in morphology and proliferation. But the cells were not germline-competent, in other words they were unable to differentiate into cells that expressed genes of the parent cells, and so they could not give rise to adult chimeras when transplanted into blastocysts. As chimeras play key roles in biomedical research, scientists identified iPSCs through a stricter gene marker that only identified iPSCS that were germline competent. It was found that cells that expressed Nanog, a gene closely tied to pluripotency, were germline competent. These cells also were virtually indistinguishable from ESCs in gene expression, and were more stable. The transgenes were better silenced in the Nanog identified cells although 20% of the iPSCs still developed tumors due to the reactivation of c-Myc. Unfortunately this stricter criterion also decreased efficiency to only 0.001-0.03%. While subsequent studies improved this efficiency by varying methods, the fact remains that iPSCs are generated with incredibly low efficiency.

iPSCs exhibit many characteristics that are related to their pluripotency. They lose proteins that are common to somatic cells and gain proteins common to embryonic cells. They also lose the G1 checkpoint in their cell cycle control mechanism, which embryonic stem cells lack as well. During the reprogramming of somatic cells in the iPS mechanism, the cell cycle structure of stem cells must be reestablished. Another distinguishing characteristic of pluripotent stem cells is their open chromatin structure, as this is needed to maintain pluripotency and to access genes rapidly for differentiation. iPSCs have the open chromatin structure associated with ESCs and other pluripotent cells. Finally, female iPSCs show reactivation of the somatically silenced X chromosome. A very early step of stem cell differentiation is the inactivation of one of the two X chromosomes in female mammals, a random process. By the reactivation of this X chromosome, iPSCs show that they are truly pluripotent and identical to ESCs.

A huge barrier to the eventual use of iPSC-derived treatments is the use of retroviruses to force the expression of the four key genes, discussed above, and activating their transcription factors. Retroviruses can carry target DNA that is inserted into a host cells genome upon injection, making them ideal for incorporating the four genes into target cells. However, this DNA and the rest of the viruses genomes remain in the host genome, which can lead to transcription of unwanted genes and greatly increases the risk of tumors. The expression of the four transgenes must be silenced after reprogramming to avoid harmful gene expression. c-Myc, a tumor-promoting gene, especially must be silenced after cellular reprogramming or the risk of tumor development becomes too great for clinical use. These retroviral methods in which the transgenes are still present in the pluripotent cells pose a danger to safety, and also are less closely related to ESCs in gene expression than their non-retroviral alternatives. Methods of reprogramming iPSCs without transgene expression in the reprogrammed cell is therefore essential not only for potential therapies and clinical applications, but also for reliable and accurate invitro models of diseases. Yet, the low efficiency of alternatives remains a worry. Whether these methods will be viable for human clinical use remains to be seen.

The excision strategy (transient transfection) of iPSC generation allows the transgenes to briefly integrate into the genome but then removes them once reprogramming is achieved. An example of this site/enzyme combination is the loxP site and the Cre enzyme. In a study of Parkinsons disease (PD), specific iPSCs, this loxP/Cre combination was used to generate the iPSCs. Neural differentiation was then induced on the iPSCs to test whether they could differentiate into dopaminergic neurons, the cells harmed in PD. The differentiation was successful, indicating the transgenes had been excised. However, a loxP site remains in iPSC genome as does some residual viral DNA, so there is still a small potential for insertional mutagenesis. The piggyBac site/enzyme system on the other hand is capable of excising itself completely, not leaving any remnants of external DNA in the iPSC genome. The piggyBac system also was much more efficient than other non-retroviral methods, with comparable efficiency to retroviral methods, but with the added benefits of safety and ease of application.

Adenoviral methods do not pose the same threats as retroviral methods of generating iPSCs. Adenoviruses work like all viruses by hijacking their hosts cellular machinery to replicate their own genome and reproduce, but unlike retroviruses they do not incorporate their genome into the host DNA. Because the transgenes are never even incorporated into the hosts genome they do not have to be excised. Instead, the genes are expressed directly from the virus genome. iPSCs created by adenoviral methods demonstrated pluripotency, but have extremely low reprogramming efficiency. Viralgenomic material could not be detected in any of the iPSCs, and no tumor formation was reported. This suggests that the use of non-integrating adenoviral methods substantially lowers the threat of tumorgenesis. The successful creation of iPSCs from adenoviral methods proves definitively that safer, non-retroviral methods can also successfully reengineer cells.

Recent studies have implied that perhaps genetic material is not required for iPS cellular reprogramming. The substitution of transgenes with small molecules that promote iPSC generation would be a safe, clinically appropriate way of creating iPSCs, though it remains to be seen if small molecules will be able to completelyreplace genetic methods of iPSC generation or are just useful as supplementary aids to the process. Protein transduction is a different method shown to entirely replace gene delivery. In this method fusion proteins are created, which fuse each of the transgenes to a cell-penetrating peptide sequence that allows it to cross the cellular membrane. Reprogramming without DNA intermediates should eliminate the risk of tumorgenesis and distorted gene expression due to the reactivation of the transgenes.

With iPSC research being a hotspot for several years now, many of the problems the technology first faced have been studied and resolved. iPSCs are now germline competent, can be generated from many different types of human and animal somatic tissue, and can be generated in a variety of retrovirus-free methods. This lack of retroviruses ended worries about transgene reactivation and subsequent tumorgenesis. The nature of the transgenes in question made the risk of tumor development particularly prevalent, as two of the genes, c-Myc and Klf4, directly inducing tumorgenesis. Retroviral delivery posed a threat to safety in its increased risk of tumorgenesis and in its tendency to alter gene expression. When other methods were established that did not require retroviruses, these concerns were put to rest, yet these new methods efficiencies must be improved and some issues still remain concerning the safety of iPSCs and their abilities to act on par with any other pluripotent cell.

Even without the use of retrovirsues, tumorgenesis is still a large concern for iPSCs, especially if they are ever to be used as cell replacement therapies. Using retroviral methods, twenty percent of iPSCs developed tumors in one study, and though this number has significantly lowered, it must become negligible for iPSCs to be considered for clinical use. It is telling that the assay for pluripotency in stem cells is the ability to form teratomas, or tumors. This test of stemness illustrates the precariously close link between stem cells and tumor cells. There are several proposals on how to prevent this tumor formation. The idea to sort cells before transplantation and after differentiation, so that only well-differentiated neural progenitors will be transplanted, is one such proposal. Another proposal is to genetically modify iPSCs so that they will have a suicide gene to self-destruct when tumors are created. Finally, some antioxidants, such as Resveratrol, have been shown to have tumor-suppressing qualities, and could potentially aid in any treatment proposed to prevent tumors (for an article about the potential of Resveratrol for the treatment of HD, click here).

Directed differentiation has been a perennial problem in stem cell biology, and iPSCs bring their own unique characteristics to the dilemma. As with ESCs, iPSCs sometimes have the tendency to not fully differentiate. Also, as with all stem cell research with neurodegenerative diseases, a more efficient and comprehensive method to differentiate cells into neural progenitors and specific neuronal tissue must be discovered, as current methods are imperfect and slow.

In iPSC research there is a need to establish methods to evaluate the reprogramming process and the final quality of the cells. To create human iPSCs suitable for cell replacement therapies, there must be tests to ensure that all pluripotent cells have differentiated, and that the cells have not been genetically altered during reprogramming or during differentiation. With cells derived from diseased individuals for an autologous treatment, there is naturally the concern that the underlying genetic cause of the disease remains in the iPSCs and will manifest itself in the same way. Some studies have indicated that iPSC lines differ drastically, which makes the reproducibility of any particular phenotype difficult. Analyzing this variability may help discover which somatic tissue is best for generating iPSCs.

A problem that has not been significantly improved upon since the beginnings of iPSC research is the technologys low efficiency. Some hypothesize that the addition of other factors would greatly aid the reprogramming process, and that reprogramming success depends on specific amounts and ratios of the four factors, which are only achieved by chance in a small percentage of the cells. Modifying the culture conditions is another area of study for increasing efficiency and rate of iPSC production. For cellular transplantation therapies, other questions must also be considered, such as the optimal cell dose and source tissue, and the best way to deliver the cells. There are potential solutions to this problem, though. Induction efficiencies have been improved up to a hundred times initial values by use of different somatic starting cells and the aid of small molecules. Although there are barriers to iPSC production, research in this field is still in its infancy and has made impressive gains for the short time it has been going on. As more studies are conducted on iPSCs, these issues may be resolved and iPSCs may enter a state capable of clinical use.

Another potential way to improve iPSC generation efficiency is to establish the best somatic cells type to reprogram for the cleanest, easiest reprogramming. Many different tissue types have been reprogrammed, including fibroblasts, neural progenitor cell, and stomach epithelial (stomach lining) cells. Certain cell types are much more efficient and rapid than others. There is also the probability that subtly varying iPSCs are generated from different types of starting tissue, some of which may prove to be useful for research or replacement purposes.

An interesting type of somatic cell was used in studies of secondary iPSCs. iPSCS were initially generated and then implanted into blastocysts to create chimeric animals. Somatic cells from these chimeras were then removed and iPSCs were generated from these cells, creating secondary iPSCs. These secondary iPSCs were generated more efficiently. The differentiation status of thecells to be reprogrammed also affects efficiency, as adult progenitor cells are reprogrammed at three hundred times the efficiency of completely differentiated somatic cells.

An interesting possibility for the reprogramming methods of iPSCs is the potential for transdifferentiation. It may not always be necessary to reprogram cells all the way back to their most primitive pluripotent stem cell state, and instead reprogram one type of adult somatic tissue directly into a different type, bypassing the lengthy processes of complete reprogramming and subsequent differentiation. For example, in theory fibroblasts that can be easily and safely obtained from a patients skin could be converted into neurons or heart muscle cells without ever passing through a pluripotent stage. This would have advantages not only in the conservation of time and resources but also for safety, as transdifferentiation does not pose the risk of tumorgenesis as the cells never are pluripotent. Unfortunately, the technology for such processes is very difficult. To reprogram cells directly into a different cell type, the qualities and characteristics of the desired cell type must be comprehensively understood. For iPSCs the desired cell type was embryonic stem cells, which were very well researched and characterized, but for many types of cell of interest, including cells of the central nervous system, there are still many unanswered questions about the target cell population. Excitingly, the Wernig lab at Stanford has recently created induced neurons (iN) directly from mouse fibroblasts.

A potential use of iPSCs for cellular therapy that can be applied much more quickly than actual replacement of damaged tissue is the transplant of pluripotent cells as support cells rather than replacement neurons. These cells offer neuroprotection by preventing inflammation and producing neurotrophic factors (for the therapeutic use of neurotrophic factors in HD, click here). In various studies, the transplantation of iPSCs has significantly improved host neuronal survival and function. This bystander mechanism of therapy is of huge immediate potential in iPSCs, and Dr. Noltas lab recently submitted a request for a clinical study of the same mechanism using mesenchymal stem cells to the FDA. For a detailed study of the use of iPSCs for this purpose click here.

Stem cell biology has been an area of great interest and intense debate since its inception, and iPSC technology has furthered this research and created hope for potential therapeutic applications. While there are still many barriers to the clinical use of stem cells, iPSCs may help elucidate the nature of both pluripotent stem cells and of many disease pathologies to reach an eventual concrete connection between the two. With their potential for autologous cell replacement and disease modeling in vitro iPSCs are the future of stem cell research, and as such they are key players in the battle against HD.

Abeliovich, Asa and Claudia A. Doege. Reprogramming Therapeutics: iPS Cell Prospects for Neurodegenerative Disease. Neuron. 12 Feb, 2009, 61 (3): 337-39.

Short, approachable article reviewing two studies deriving iPSCs from patients with neurological disorders.

Cox, Jesse L. and Angie Rizzino. Induced pluripotent stem cells: what lies beyond the paradigm shift. Experimental Biology and Medicine. Feb 2010, 235 (2): 148-58.

Very detailed, mostly accessible review of the state of iPS research and the discoveries to date, as well as what iPS cells mean for stem cell biology and modern medical approaches. Perfect thorough introduction to iPS technology.

Crook, Jeremy Micah, and Nao Rei Kobayashi. Human stem cells for modeling neurological disorders: Accelerating the drug discovery pipeline. Journal of Cellular Biochemistry. 105 (6): 1361-66.

Accessible, interesting article that argues the greatest potential for iPSCs is to test potential drugs for neurological diseases in vitro and find problems early on in the drug development, saving time and resources.

Gunaseeli, I., et al. Induced Pluripotent Stem Cells as a Model for Accelerated Patient- and Disease-specific Drug Discovery. Current Medicinal Chemistry. 2010, 17: 759-766.

Readable review on the future of iPS cells, comparing them with other stem cells and elucidating their pontential drawbacks. Good summary of the landmark discoveries in iPS technology to date.

Haruhisa, Inoue. Neurodegenerative disease-specific induced pluripotent stem cell research. Experimental Cell Research. 2010.

General overview of use of iPS cells specific to neurological diseases for modeling diseases in vitro and eventually using as a cellular replacement therapy. Good, non-technical overview of the various potential pathways of iPS technology.

Hung, Chia-Wei, et al. Stem Cell-Based Neuroprotective and Neurorestorative Strategies. International Journal of Molecular Science. 2010, 11(5): 20392055.

Overview of various neurological diseases and the potential of stem cell therapeutics, either using adult neural stem cells or iPS stem cells. Experiment descriptions are fairly technical, but the reviews reflections and discussion are accessible and interesting.

Laowtammathron, Chuti, et al. Monkey hybrid stem cells develop cellular features of Huntingtons disease. BioMed Center Cell Biology. 2010, 11 (12).

Detailed article on the establishment of pluripotent HD monkey model cell line and its use in the study of Huntingtons.

Marchetto, Maria C.N., et al. Pluripotent stem cells in neurodegenerative and neurodevelopmental diseases. Human Molecular Genetics. 2010, 19 (1).

Fairly technical review describing the use of iPSCs for modeling neurological disorders.

Niclis, J.C., et al. Human embryonic stem cell models of Huntingtons Disease. Reproductive Biomedicine Online. July 2009, 19 (1): 106-13.

Detailed, technical article on the use of human embryonic stem cell lines for HD.

OMalley, James. New strategies to generate induced pluripotent stem cells. Current Opinions in Biotechnology. Oct. 2009: 20 (5): 516-21.

Longer technical article on the various strategies to generate iPS cells without using potentially dangerous viral vectors.

Okita, Keisuke, et al. Generation of germline-competent induced pluripotent stem cells. Nature. 19 Jul, 2007, 448(7151):313-17.

Fairly technical article about an early study in iPS research, where cells were selected for Nanog expression rather than the less pertinent gene Fbx15. This higher caliber of selected cells were germline-competent.

Okita, Keisuke, et al. Generation of Mouse Induced Pluripotent Stem Cells Without Viral Vectors. Science. 7 Nov, 2008, 322 (5903): 949-53.

Technical article about the advancements in finding non-viral, clinically applicable methods of creating iPS cells.

Orlacchio, A., et al. Stem Cells: An Overview of the Current Status of Therapies for Central and Peripheral Nervous System Diseases. Current Medicinal Chemistry. 2010, 17: 595-608.

Technical review on the various types of stem cells used in the studies of neurological diseases and the progress made to date with these cells.

Park, In-Hyun, et al. Disease-Specific Induced Pluripotent Stem Cells. Cell. 2008, 134 (5): 877-86.

Fairly accessible article on the creation of iPS cells with genetic defects, as tools for studying the symptoms and experimenting with treatments of various diseases.

Robbins, Reisha D., et al. Inducible pluripotent stem cells: not quite ready for prime time? Current Opinion in Organ Transplantation. 15 (1): 61-57.

Clear review of the barriers facing clinical use of iPSCs, accessible and realistic.

Soldner, Frank, et al. Parkinsons Disease Patient-Derived Induced Pluripotent Stem Cells Free of Viral Reprogramming Factors. Cell. 6 Mar, 2009, 136 (5): 964-77.

Technical article about first successful derivation of iPS cells from a patient with a neurodegenerative disease without using viral vectors. Relevant to HD research as a protocol that will likely be followed for subsequent creation of neurodegenerative iPSC lines for in vitro study.

Stradtfeld, Matthias, et al. Induced Pluripotent Stem Cells Generated Without Viral Integration. Science. 7 Nov, 2008, 322 (5903): 945-49.

Technical article outlining a method for creating iPS cells using excisable adenoviruses, rather. than retroviruses that have the potential to harm the cells.

Takahashi, Kazutoshi, et al. Induction of Pluripotent Stem Cells from Adult Human Fibroblasts by Defined Factors Cell. 30 Nov, 2007, 131(5): 861-72.

Landmark article in the discovery of induced pluripotent stem cells and the factors that create them. Short, but fairly technical.

Yamanaka, Shinya. Induction of pluripotent stem cells from mouse fibroblasts by four transcription factors. Cell Proliferation. Feb, 2008, 41 (Suppl. 1):51-6

Short review, less technical summary of first iPS discovery by Yamanaka. Perfect for quick overview of the basics of iPS cell generation.

Yamanaka, Shinya. Strategies and New Developments in the Generation of Patient-Specific Pluripotent Stem Cells. Cell: Stem Cell. 7 June 2007, 1(1): 39-49.

Comprehensive review of various methods for creating pluripotent stem cells with a detailed introduction to iPSC methods. Fairly accessible, and very thorough.

A. Lanctot 2011

The rest is here:
Induced Pluripotent Stem Cells: The Future of Tissue ...