Category Archives: Induced Pluripotent Stem Cells


The stem cell assays market is projected to reach USD 4.5 Billion by 2027 from USD 1.9 Billion in 2022, at a CAGR of 17.7% – Yahoo Finance

ReportLinker

during the forecast period. The growth of the market is projected to be driven by collaborations and agreements among market players for stem cell assay products & services, launch of new stem cell analysis systems such as flow cytometers and increase in R&D expenditure by biopharmaceutical and biotechnology companies.

New York, June 28, 2022 (GLOBE NEWSWIRE) -- Reportlinker.com announces the release of the report "Stem Cell Assays Market by Type, Cell Type, Product & Service, Application, End User - Global Forecast to 2027" - https://www.reportlinker.com/p04221306/?utm_source=GNW

The viability/cytotoxicity assays accounted for the largest share of the type segment in the stem cell assays market in 2021. Cell viability assays help to determine the number of live and dead cells in a culture medium.The viability/cytotoxicity assays includes various types such as tetrazolium reduction assays, resazurin cell viability assays, calcein-AM cell viability assays, and other viability/cytotoxicity assays.

The cell viability/cytotoxicity market is likely to be driven by rise R&D spending on stem cell research, increase in demand for stem cell assays in drug discovery and development of new stem cell therapies..

The adult stem cells segment accounted for the largest share of the cell type segment in the stem cell assays market in 2021. The adult stem cells account for the largest share of the stem cell assays market.The adult stem cells include mesenchymal stem cells, induced pluripotent stem cells, hematopoietic stem cells, umbilical cord stem cells, and neural stem cells.

The growth of adult stems cells segment is driven by increasing usage of adult stem cells in regenerative medicine and development of advanced therapies. Key players in the stem cell assays market include Thermo Fisher Scientific Inc. (US), Merck KGaA (Germany), Danaher (US), Becton, Dickinson and Company (US), Bio-Rad Laboratories (US), PerkinElmer (US), Agilent Technologies (US), Promega Corporation (US), Cell Biolabs (US), Miltenyi Biotec (Germany), STEMCELL Technologies (Canada), Bio-Techne Corporation (US), FUJIFILM Holdings Corporation (Japan), Charles River Laboratories (US), HemoGenix Inc. (US), Lonza Group (Switzerland), Takara Bio Inc. (Japan), Creative Bioarray (US), AAT Bioquest, Inc. (US), BPS Bioscience, Inc. (US), Enzo Biochem (US), PromoCell GmbH (Germany), Biotium (US), Geno Technology (US), Abcam plc (UK), and ReachBio Research Labs (US).

Asia Pacific: The fastest-growing region in the stem cell assays market The Asia Pacific is estimated to be the fastest-growing segment of the market, owing to rising prevalence of cancer & other diseases, increasing R&D spending on biopharmaceutical projects and focus on developing stem cell based therapies. In this region, China and Japan are the largest markets.

The primary interviews conducted for this report can be categorized as follows: By Respondent: Supply Side- 80% and Demand Side - 20% By Designation: Managers- 45%, CXOs and Director level - 30%, and Executives - 25% By Region: North America -20%, Europe -10%, Asia-Pacific -55%, RoW -15%

List of Companies Profiled in the Report: Thermo Fisher Scientific Inc. (US) Merck KGaA (Germany) Danaher (US) Becton, Dickinson and Company (US) Bio-Rad Laboratories (US) PerkinElmer (US) Agilent Technologies (US) Promega Corporation (US) Cell Biolabs (US) Miltenyi Biotec (Germany) STEMCELL Technologies (Canada) Bio-Techne Corporation (US) FUJIFILM Holdings Corporation (Japan) Charles River Laboratories (US) HemoGenix Inc. (US) Lonza Group (Switzerland) Takara Bio Inc. (Japan) Creative Bioarray (US) AAT Bioquest, Inc. (US) BPS Bioscience, Inc. (US) Enzo Biochem (US) PromoCell GmbH (Germany) Biotium (US) Geno Technology (US) Abcam plc (UK) ReachBio Research Labs (US).

Research Coverage: This report provides a detailed picture of the stem cell assays market.It aims at estimating the size and future growth potential of the market across different segments such as the product, functionality, formulation and region.

The report also includes an in-depth competitive analysis of the key market players along with their company profiles recent developments and key market strategies.

Key Benefits of Buying the Report: The report will help market leaders/new entrants by providing them with the closest approximations of the revenue numbers for the overall stem cell assays market and its subsegments.It will also help stakeholders better understand the competitive landscape and gain more insights to better position their business and make suitable go-to-market strategies.

This report will enable stakeholders to understand the markets pulse and provide them with information on the key market drivers, restraints, challenges, trends, and opportunities. Read the full report: https://www.reportlinker.com/p04221306/?utm_source=GNW

About Reportlinker ReportLinker is an award-winning market research solution. Reportlinker finds and organizes the latest industry data so you get all the market research you need - instantly, in one place.

__________________________

Story continues

Read the original here:
The stem cell assays market is projected to reach USD 4.5 Billion by 2027 from USD 1.9 Billion in 2022, at a CAGR of 17.7% - Yahoo Finance

Fate Therapeutics Announces Expansion of Solid Tumor Collaboration with ONO Pharmaceutical for Off-the-Shelf, iPSC-derived CAR NK and CAR T-Cell…

Fate Therapeutics, Inc.

ONO to Contribute Novel Binding Domains for a Second Solid Tumor Antigen to the Collaboration

Expanded Partnership Enables Development of both CAR NK and CAR T-cell Collaboration Candidates for Solid Tumors

SAN DIEGO, June 28, 2022 (GLOBE NEWSWIRE) -- Fate Therapeutics, Inc. (NASDAQ: FATE), a clinical-stage biopharmaceutical company dedicated to the development of programmed cellular immunotherapies for patients with cancer, today announced that it has expanded its off-the-shelf, iPSC-derived, cell-based cancer immunotherapy collaboration with ONO Pharmaceutical Co., Ltd. (ONO) to include the development of chimeric antigen receptor (CAR) NK cell collaboration candidates. In addition, as part of the collaborations expansion, ONO will contribute novel binding domains targeting a second solid tumor antigen. Under the original Collaboration and Option Agreement entered into between Fate and ONO in September 2018, ONO has contributed novel binding domains targeting an initial solid tumor antigen, and Fate is currently conducting preclinical development of a multiplexed-engineered, iPSC-derived CAR T-cell product candidate for solid tumors.

Our collaboration with ONO has focused on driving innovation in the field of cell therapy for solid tumors, and we are excited by the preclinical data we have observed with our first iPSC-derived CAR T-cell product candidate, said Scott Wolchko, President and Chief Executive Officer of Fate Therapeutics. We are impressed with the differentiated antigen binders that ONO has contributed to the partnership, and we are pleased to expand our collaboration to initiate preclinical development of collaboration products targeting a second solid tumor antigen.

Under the terms of the amended Collaboration and Option Agreement, Fate will advance iPSC-derived CAR NK and CAR T-cell product candidates to a pre-defined preclinical milestone, at which point ONO has an option to assume responsibility for worldwide development and commercialization with Fate retaining the right to jointly develop and commercialize in the United States and Europe. Fate retains all rights of manufacture of collaboration products on a global basis.

Story continues

The first multiplexed-engineered, iPSC-derived CAR T-cell product candidate under our collaboration with Fate Therapeutics incorporates multiple mechanisms of action designed to specifically address solid tumors and is successfully advancing toward clinical development, said Toichi Takino, Senior Executive Officer / Executive Director, Discovery & Research of ONO. Based on the collaboration progress and Fates proven ability to develop innovative product candidates, we are excited to expand our collaboration to include a second solid tumor target and to continue our work with Fate in developing first-in-class, off-the-shelf CAR NK and CAR T-cell therapies for cancer patients.

Fate will continue to receive committed research funding from ONO during the preclinical option period, and is eligible to receive a preclinical option exercise fee as well as clinical, regulatory and commercialization milestone payments from ONO in connection with the development and commercialization of each product candidate. In addition, Fate is eligible to receive tiered royalties on net sales by ONO of each product candidate in the ONO territory.

About Fate Therapeutics iPSC Product Platform The Companys proprietary induced pluripotent stem cell (iPSC) product platform enables mass production of off-the-shelf, engineered, homogeneous cell products that are designed to be administered with multiple doses to deliver more effective pharmacologic activity, including in combination with other cancer treatments. Human iPSCs possess the unique dual properties of unlimited self-renewal and differentiation potential into all cell types of the body. The Companys first-of-kind approach involves engineering human iPSCs in a one-time genetic modification event and selecting a single engineered iPSC for maintenance as a clonal master iPSC line. Analogous to master cell lines used to manufacture biopharmaceutical drug products such as monoclonal antibodies, clonal master iPSC lines are a renewable source for manufacturing cell therapy products which are well-defined and uniform in composition, can be mass produced at significant scale in a cost-effective manner, and can be delivered off-the-shelf for patient treatment. As a result, the Companys platform is uniquely designed to overcome numerous limitations associated with the production of cell therapies using patient- or donor-sourced cells, which is logistically complex and expensive and is subject to batch-to-batch and cell-to-cell variability that can affect clinical safety and efficacy. Fate Therapeutics iPSC product platform is supported by an intellectual property portfolio of over 350 issued patents and 150 pending patent applications.

About Fate Therapeutics, Inc. Fate Therapeutics is a clinical-stage biopharmaceutical company dedicated to the development of first-in-class cellular immunotherapies for patients with cancer. The Company has established a leadership position in the clinical development and manufacture of universal, off-the-shelf cell products using its proprietary induced pluripotent stem cell (iPSC) product platform. The Companys immuno-oncology pipeline includes off-the-shelf, iPSC-derived natural killer (NK) cell and T-cell product candidates, which are designed to synergize with well-established cancer therapies, including immune checkpoint inhibitors and monoclonal antibodies, and to target tumor-associated antigens using chimeric antigen receptors (CARs). Fate Therapeutics is headquartered in San Diego, CA. For more information, please visit http://www.fatetherapeutics.com.

Fate Therapeutics Forward-Looking Statements This release contains "forward-looking statements" within the meaning of the Private Securities Litigation Reform Act of 1995, including statements regarding the impact, timing, conduct and the potential benefits of the collaboration, including expected funding and payments to be received by Fate Therapeutics under the collaboration, as well as the capabilities, expertise and responsibilities of each of Fate Therapeutics and ONO Pharmaceutical. These and any other forward-looking statements in this release are based on management's current expectations of future events and are subject to a number of risks and uncertainties that could cause actual results to differ materially and adversely from those set forth in or implied by such forward-looking statements. These risks and uncertainties include, but are not limited to, risks associated with: the success, cost and timing of product development activities under the collaboration; the ability of Fate Therapeutics and ONO Pharmaceutical to obtain regulatory approval for and to commercialize any product candidates developed under the collaboration; regulatory requirements and regulatory developments; the success of competing treatments and technologies; the risk of cessation or delay of any development activities under the collaboration for a variety of reasons; any adverse effects or events, or other negative results, that may be observed in preclinical or clinical development of any product candidates developed through the collaboration; and the risk that funding and payments received by Fate Therapeutics under the collaboration may be less than expected. For a discussion of other risks and uncertainties, and other important factors, any of which could cause Fate Therapeutics actual results to differ from those contained in the forward-looking statements, see the risks and uncertainties detailed in Fate Therapeutics periodic filings with the Securities and Exchange Commission, including but not limited to Fate Therapeutics most recently filed periodic report, and from time to time in Fate Therapeutics press releases and other investor communications.Fate Therapeutics is providing the information in this release as of this date and, except as required by law, does not undertake any obligation to update any forward-looking statements contained in this release as a result of new information, future events or otherwise.

Contact: Christina Tartaglia Stern Investor Relations, Inc. 212.362.1200 christina@sternir.com

Go here to read the rest:
Fate Therapeutics Announces Expansion of Solid Tumor Collaboration with ONO Pharmaceutical for Off-the-Shelf, iPSC-derived CAR NK and CAR T-Cell...

Human iPS Cell Characterization Kit Market to Garner Brimming Revenues with Comprehensive Analysis and Landscape Outlook to 2028- Merck KGaA, Takara…

The Human iPS Cell Characterization Kit (Induced pluripotent stem cell) contains a range of sensitive tools for the phenotypic assessment of the pluripotent status of human Embryonic stem & induced pluripotent Stem cells.

This report studies the Human iPS Cell Characterization Kit Market with many aspects of the industry like the market size, market status, market trends and forecast, the report also provides brief information of the competitors and the specific growth opportunities with key market drivers. Find the complete Human iPS Cell Characterization Kit Market analysis segmented by companies, region, type and applications in the report.

The report offers valuable insight into the Human iPS Cell Characterization Kit market progress and approaches related to the Human iPS Cell Characterization Kit market with an analysis of each region. The report goes on to talk about the dominant aspects of the market and examine each segment.

Key Players: Merck KGaA, Takara Bio, Thermo Fisher Scientific, BD Biosciences, Applied StemCell, Amsbio, Bio-Techne, ALSTEM, STEMCELL Technologies, System Biosciences, Applied Biological Materials, Creative Bioarray, Elixirgen Scientific, and Miltenyi Biotec

Get Sample Copy @ https://www.reportsandmarkets.com/sample-request/global-human-ips-cell-characterization-kit-market-4457996?utm_source=dj&utm_medium=34

The global Human iPS Cell Characterization Kit market is segmented by company, region (country), by Type, and by Application. Players, stakeholders, and other participants in the global Human iPS Cell Characterization Kit market will be able to gain the upper hand as they use the report as a powerful resource. The segmental analysis focuses on revenue and forecast by region (country), by Type, and by Application for the period 2022-2026.

Market Segment by Regions, regional analysis covers

North America (United States, Canada and Mexico)

Europe (Germany, France, UK, Russia and Italy)

Asia-Pacific (China, Japan, Korea, India and Southeast Asia)

South America (Brazil, Argentina, Colombia etc.)

Middle East and Africa (Saudi Arabia, UAE, Egypt, Nigeria and South Africa)

Research objectives:

To study and analyze the global Human iPS Cell Characterization Kit market size by key regions/countries, product type and application, history data from 2013 to 2017, and forecast to 2027.

To understand the structure of Human iPS Cell Characterization Kit market by identifying its various sub segments.

Focuses on the key global Human iPS Cell Characterization Kit players, to define, describe and analyze the value, market share, market competition landscape, SWOT analysis and development plans in next few years.

To analyze the Human iPS Cell Characterization Kit with respect to individual growth trends, future prospects, and their contribution to the total market.

To share detailed information about the key factors influencing the growth of the market (growth potential, opportunities, drivers, industry-specific challenges and risks).

To project the size of Human iPS Cell Characterization Kit submarkets, with respect to key regions (along with their respective key countries).

To analyze competitive developments such as expansions, agreements, new product launches and acquisitions in the market.

To strategically profile the key players and comprehensively analyze their growth strategies.

The report lists the major players in the regions and their respective market share on the basis of global revenue. It also explains their strategic moves in the past few years, investments in product innovation, and changes in leadership to stay ahead in the competition. This will give the reader an edge over others as a well-informed decision can be made looking at the holistic picture of the market.

Key questions answered in this report

What will the market size be in 2027 and what will the growth rate be?

What are the key market trends?

What is driving this market?

What are the challenges to market growth?

Who are the key vendors in this market space?

What are the market opportunities and threats faced by the key vendors?

What are the strengths and weaknesses of the key vendors?

Table of Contents: Human iPS Cell Characterization Kit Market

Chapter 1: Overview of Human iPS Cell Characterization Kit Market

Chapter 2: Global Market Status and Forecast by Regions

Chapter 3: Global Market Status and Forecast by Types

Chapter 4: Global Market Status and Forecast by Downstream Industry

Chapter 5: Market Driving Factor Analysis

Chapter 6: Market Competition Status by Major Manufacturers

Chapter 7: Major Manufacturers Introduction and Market Data

Chapter 8: Upstream and Downstream Market Analysis

Chapter 9: Cost and Gross Margin Analysis

Chapter 10: Marketing Status Analysis

Chapter 11: Market Report Conclusion

Chapter 12: Research Methodology and Reference

Make an enquiry before buying this report @ https://www.reportsandmarkets.com/sample-request/global-human-ips-cell-characterization-kit-market-4457996?utm_source=dj&utm_medium=34

About Us:

Our marketing research reports comprise of the best market analysis along with putting the right statistical and analytical information on the markets, applications, industry analysis, market shares, technology and technology shifts, important players, and the developments in the market. If you require any specific company, then our company reports collection has countless profiles of all the key industrial companies. All these reports comprise of vital information including the company overview, the company history, the business description, the key products & services, the SWOT analysis, the crucial facts, employee details, the locations and subsidiaries to name a few.

Contact Us:

Sanjay Jain

Manager Partner Relations & International Marketing

http://www.reportsandmarkets.com

Ph: +1-352-353-0818 (US)

The post Human iPS Cell Characterization Kit Market to Garner Brimming Revenues with Comprehensive Analysis and Landscape Outlook to 2028- Merck KGaA, Takara Bio, Thermo Fisher Scientific, BD Biosciences appeared first on Agency.

Read more here:
Human iPS Cell Characterization Kit Market to Garner Brimming Revenues with Comprehensive Analysis and Landscape Outlook to 2028- Merck KGaA, Takara...

Sernova and Evotec Enter into an Exclusive Global Strategic Partnership for iPSC-Based Beta Cell Replacement Therapy to Develop and Commercialize a…

LONDON, Ontario, May 17, 2022 (GLOBE NEWSWIRE) -- Sernova Corp. (TSX-V:SVA)(

The collaboration agreement is a transformative partnership that combines Sernovas Cell Pouch System technologies, which has demonstrated Phase 1/2 clinical proof-of-concept using human donor islets, with Evotecs iPSC-based beta cells. Incorporating Evotecs insulin-producing, ethically-derived beta cells within Sernovas Cell Pouch platform creates the potential to provide a functional cure for millions of people suffering from diabetes using a cGMP controlled and scalable off-the-shelf product.

With its long-standing beta cell development program, Evotec has demonstrated the ability to reliably produce an unlimited supply of high quality, stable, human iPSC-derived beta cells using its proprietary process for producing islet-like clusters in a quality-controlled scalable bioreactor process. These islet-like clusters have now been proven to be functionally equivalent to primary human islets in their ability to normalize blood glucose using in vivo models of type 1 diabetes (T1D).

The partnership provides Sernova a global exclusive option to license Evotecs iPSC-based beta cells for use in treating both type 1 and type 2 diabetes. In addition to entering into the collaboration agreement, Evotec has made a strategic equity investment of 15M and will make a further investment of 5M. Specifically, concurrently with the entering into the collaboration agreement, Evotec acquired a total of 12,944,904 common shares at a price of CAD $1.57 per share for gross proceeds to Sernova of $20,323,500. In addition, pursuant to an unconditional purchase warrant, Evotec will acquire, on or before August 31, 2022, a further 2,709,800 common shares at a price of CAD$2.50 per share for gross proceeds of $6,774,500. All of the securities issued to Evotec are subject to a four month hold period.

Further to the collaboration and Evotecs strategic equity investment, Dr. Cord Dohrmann, Chief Scientific Officer of Evotec will join Sernovas Board of Directors.

Dr. Philip Toleikis, President, and Chief Executive Officer of Sernova, commented, In tandem with our current clinical islet cell program, Sernova entered into multiple pharmaceutical research collaborations to identify the highest quality and most compatible iPSC cell technology, and validate the cells preclinically within our Cell Pouch System. Evotec is an iPSC powerhouse having dedicated many years and substantial resources to developing high quality and stable stem cell technologies for multiple therapeutic applications. In every sense, both as a global strategic partner and as an iPSC expert, Evotec has exceeded all our expectations and Dr. Dohrmanns appointment to Sernovas Board adds significant regenerative medicine depth and cell therapy expertise. Todays announcement of this joint iPSC beta-cell partnership completes the three pillars of our diabetes cell therapy platform. Alongside our clinically validated Cell Pouch System and recently acquired conformal coating immune protection technology, this now establishes a total regenerative medicine cell therapy solution for insulin-dependent diabetes.

Dr. Cord Dohrmann, Chief Scientific Officer of Evotec, commented, We searched long and hard for the right partner. Sernova clearly ticks all boxes with their clinically validated Cell Pouch technology, which fits perfectly to Evotecs iPSC-based beta cells. Together we will progress a highly differentiated first-in-class beta cell therapy into clinical development with the common goal to bring a truly transformative therapy to insulin-dependent diabetic patients. The synergies of Evotecs and Sernovas technologies puts Sernova in position to become the worlds leader in beta cell replacement therapy. Our equity investment underlines our strategic interest in this collaboration with Sernova. I am very much looking forward to collaborating with Sernova on the project as well as contributing to their Board of Directors.

Sernova has acquired an option for an exclusive global license to Evotecs Induced Pluripotent Stem Cell (iPSC)-based Beta cells to treat patients with insulin-dependent diabetes. From an operational perspective, the preclinical development program(s) will be jointly funded by Sernova and Evotec until IND acceptance. Sernova has the right to exercise its option for an exclusive global license upon IND filing. Evotec will contribute its cell manufacturing capabilities through research, development and product commercialization and will decide in the future on the joint funding of clinical development. Upon commercialization, there will be a profit-sharing arrangement between the two companies, with the split being dependent on Evotecs participation in funding the clinical development program.

Joint Sernova / Evotec Conference Call and Webcast Details:

Date: Tuesday, May 17, 2022 Time: 8:30 am EDT US Toll Free: 1-877-704-4453 International: 1-201-389-0920 Conference ID: 13730121 Webcast: https://viavid.webcasts.com/starthere.jsp?ei=1550130&tp_key=3de87cce1d

A simultaneous slide presentation will be available via the above webcast link.

ABOUT SERNOVA CORP AND THE CELL POUCH SYSTEM CELL THERAPY PLATFORM

Sernova Corp is developing regenerative medicine therapeutic technologies using a medical device and immune protected therapeutic cells (i.e., human donor cells, corrected human cells and stem-cell derived cells) to improve the treatment and quality of life of people with chronic metabolic diseases such as insulin- dependent diabetes, blood disorders including hemophilia, and other diseases treated through replacement of proteins or hormones missing or in short supply within the body.

The Cell Pouch, as part of the Cell Pouch System, is a proprietary, scalable, implantable macro- encapsulation device solution designed for the long-term survival and function of therapeutic cells. After implantation, the device incorporates with tissue, forming highly vascularized, native tissue chambers for the transplantation and function of therapeutic cells, that release proteins and hormones as required to treat disease.

The Cell Pouch, along with therapeutic cells, has been shown to provide long-term safety and efficacy in small and large animal models of diabetes and has been proven to provide a biologically compatible environment for insulin-producing cells in humans in a Canadian first-in-human study. Sernova is currently conducting a Phase 1/2 clinical trial study at the University of Chicago. Encouraging interim results have been presented at several international scientific conferences.

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

ABOUT EVOTEC AND iPSC

Induced pluripotent stem cells (also known as iPS cells or iPSCs) are a type of pluripotent stem cell that can be generated directly from adult cells. The iPSC technology was pioneered by Shinya Yamanakas lab in Kyoto, Japan, who showed in 2006 that the introduction of four specific genes encoding transcription factors could convert adult cells into pluripotent stem cells. He was awarded the 2012 Nobel Prize along with Sir John Gurdon for the discovery that mature cells can be reprogrammed to become pluripotent. Pluripotent stem cells hold great promise in the field of regenerative medicine. Because they can propagate indefinitely, as well as give rise to every other cell type in the body (such as neurons, heart, pancreatic and liver cells), they represent a single source of cells that could be used to replace those lost to damage or disease.

Evotec has built an industrialised iPSC infrastructure that represents one of the largest and most sophisticated iPSC platforms in the industry. Evotecs iPSC platform has been developed over the last years with the goal to industrialise iPSC-based drug screening in terms of throughput, reproducibility and robustness to reach the highest industrial standards, and to use iPSC-based cells in cell therapy approaches via the Companys proprietary EVOcells platform.

For further information contact:

Corporate and Investors: Sernova Corp Christopher Barnes Tel: (519) 858-5126 [emailprotected] http://www.sernova.com

Investors: Corey Davis, Ph.D. LifeSci Advisors, LLC [emailprotected] Tel: 212-915-2577

Media: Elizabeth Miller, MD LifeSci Communications [emailprotected]

FORWARD-LOOKING INFORMATION

This release may contain forward-looking statements. Forward-looking statements are statements that are not historical facts and are generally, but not always, identified by the words expects, plans, anticipates, believes, intends, estimates, projects, potential and similar expressions, or that events or conditions will, would, may, could or should occur. Although Sernova believes the expectations expressed in such forward-looking statements are based on reasonable assumptions, such statements are not guarantees of future performance, and actual results may differ materially from those in forward-looking statements. Forward-looking statements are based on the beliefs, estimates, and opinions of Sernovas management on the date such statements were made, which include our beliefs about the conduct and outcome of clinical trials, and the development of new technologies, cell therapy solutions and or products. The information disclosed represents results from one patient and may not be representative of all study patients or of the final study results. Sernova expressly disclaims any intention or obligation to update or revise any forward-looking statements whether as a result of new information, future events or otherwise.

Read the original:
Sernova and Evotec Enter into an Exclusive Global Strategic Partnership for iPSC-Based Beta Cell Replacement Therapy to Develop and Commercialize a...

Stem Cell Antibody Market Detailed Strategies, Competitive Landscaping and Developments for next 5 years Indian Defence News – Indian Defence News

Stem cell antibodies are the specialized cells which have the capacity to self-renew and differentiate to the specialized cell type such as neurons, liver and muscle cells. The Adult stem cells such as neural stem cells are multi potent. These have the ability to maintain and repair tissues where they are found. It has various applications such as developmental biology, disease modeling research, drug development screening and cell therapy studies. The methods of antibody detection such as immunocytochemistry and flow cytometry are most commonly used for stem cells and differentiated cell characterizations.

The latest study released on the Global Stem Cell Antibody Market by AMA Research evaluates market size, trend, and forecast to 2027. The Stem Cell Antibody market study covers significant research data and proofs to be a handy resource document for managers, analysts, industry experts and other key people to have ready-to-access and self-analyzed study to help understand market trends, growth drivers, opportunities and upcoming challenges and about the competitors.

Key Players in This Report Include:

Thermo Fisher Scientific, Inc. (United States), Merck Group (Germany), Abcam plc (United Kingdom), Becton, Dickinson and Company (United States), Bio-Rad Laboratories, Inc. (United States), Cell Signaling Technology, Inc. (United States), Agilent Technologies, Inc. (United States), F. Hoffmann-La Roche Ltd (Switzerland), Danaher Corporation (United States), GenScript (United States),

Download Sample Report PDF (Including Full TOC, Table & Figures) @ https://www.advancemarketanalytics.com/sample-report/48964-global-stem-cell-antibody-market

Market Drivers:

Market Opportunities:

Market Trend:

The Global Stem Cell Antibody Market segments and Market Data Break Down are illuminated below:

by Type (Primary Antibodies, Secondary Antibodies), Application (Developmental biology, Disease modeling research, Drug development screening, Cell therapy studies), Stem cell type (Adult stem cells, Embryonic (or pluripotent) stem cells, Induced pluripotent stem cells), Methods of detection (Immunocytochemistry, Flow cytometry)

Global Stem Cell Antibody market report highlights information regarding the current and future industry trends, growth patterns, as well as it offers business strategies to help the stakeholders in making sound decisions that may help to ensure the profit trajectory over the forecast years.

Have a query? Market an enquiry before purchase @ https://www.advancemarketanalytics.com/enquiry-before-buy/48964-global-stem-cell-antibody-market

Geographically, the detailed analysis of consumption, revenue, market share, and growth rate of the following regions:

Objectives of the Report

Buy Complete Assessment of Stem Cell Antibody market Now @ https://www.advancemarketanalytics.com/buy-now?format=1&report=48964

Major highlights from Table of Contents:

Stem Cell Antibody Market Study Coverage:

Browse Complete Summary and Table of Content @ https://www.advancemarketanalytics.com/reports/48964-global-stem-cell-antibody-market

Key questions answered

Contact US:

Craig Francis (PR & Marketing Manager)

AMA Research & Media LLP

Unit No. 429, Parsonage Road Edison, NJ

New Jersey USA 08837

Continued here:
Stem Cell Antibody Market Detailed Strategies, Competitive Landscaping and Developments for next 5 years Indian Defence News - Indian Defence News

Cell Expansion Market Analytical Overview, COVID-19 Impact Analysis, Growth Factors, Demand and Trends Forecast 2022 to 2030 Designer Women -…

Decision Foresight is working on new industry report named, Cell Expansion Market 2020-2030. According to the Decision Foresight, Cell Expansion Market held USD 16.68 Bn in 2021 and is to grow with a CAGR of 14.3% from 2022-2030.

The Decision Foresight is dedicated to provide you with an in-depth analysis of the various market based on its verticals, manufacturing process, products types, applications, end-users and regional analysis which further bifurcated into country level study. The study we provide covered the insightful data about all the channels related to your domain. In addition, the elaborated study of key market players involved in Cell Expansion Market gives predicted size of the market and opportunities in future.

Get sample of this report @ https://www.decisionforesight.com/request-sample/CUS020993

Market Segments:

The key segments covered in the report are:

Regional Analysis

Detailed regional and country-wise analysis, trend, forecasts has been covered in the report. The countries covered in the regional analysis of the report are (U.S., Canada, Mexico) in North America, (U.K., Germany, France, Italy, Spain, and Rest of Europe) in Europe, (China, India, Japan, Singapore, Malaysia, Australia, South Korea, Rest of Asia-Pacific (APAC)) in the Asia-Pacific (APAC), (Saudi Arabia, U.A.E, South Africa, Turkey, Rest of Middle East and Africa (MEA) as a part of Middle East and Africa (MEA), and (Argentina, Brazil, and Rest of South America) as part of South America.

Market Report Scope

To Enquiry before Buy this Research Report or For Any Customization Ask:

@ https://www.decisionforesight.com/inquire/CUS020993

Key Players:

Global Cell Expansion Market report covers prominent players like thermo fisher scientific, inc. (us), danaher (us), becton, dickinson and company (us), lonza (switzerland), corning, inc. (us), merck kgaa (germany), sartorius stedim biotech (france), getinge ab (sweden) terumo corporation (japan), and miltenyi biotec (germany)

Key Points covered in the report:

Ask Industry Expert for Any Specific Query

@ https://www.decisionforesight.com/speak-analyst/CUS020993

Key questions answered in the report:

Purchase this Market Research Report Now @ https://www.decisionforesight.com/checkout/CUS020993

About US

Decision Foresight is a new-age market research company with a firm grip on the pulse of global markets. Decision Foresight Markets has emerged as a dependable source for the market research needs of businesses within a quick time span. We have collaborated with leading publishers of market intelligence and the coverage of our reports reserve spans all the key industry verticals and thousands of micro markets. The massive repository allows our clients to pick from recently published reports from a range of publishers that also provide extensive regional and country-wise analysis. Moreover, pre-booked research reports are among our top offerings.

The collection of market intelligence reports is regularly updated to offer visitors ready access to the most recent market insights. We provide round-the-clock support to help you repurpose search parameters and thereby avail a complete range of reserved reports. After all, it is all about helping you reach an informed strategic decision about purchasing the right report that caters to all your market research demands.

Read More Reports:

Anaesthesia Face Masks Market:

https://www.decisionforesight.com/reports/anaesthesia-face-masks-market

Anaesthesia Monitoring Devices Market:

https://www.decisionforesight.com/reports/anaesthesia-monitoring-devices-market

Analgesics Market:

https://www.decisionforesight.com/reports/analgesics-market

Angiography Devices Market:

https://www.decisionforesight.com/reports/angiography-devices-market

Animal Health Market:

https://www.decisionforesight.com/reports/animal-health-market

Contact Us

Aloke Kumar,

Global Business Development Manager,

Decision Foresight

http://www.decisionforesight.com

Phone: +919875577842

Email: sales@decisionforesight.com;

aloke@decisionforesight.com

View original post here:
Cell Expansion Market Analytical Overview, COVID-19 Impact Analysis, Growth Factors, Demand and Trends Forecast 2022 to 2030 Designer Women -...

Technical Advancements & Innovative Products Likely to Expand Application of Surgical Meshes in Untapped Domains, States Fact.MR – BioSpace

Global Surgical Mesh Market Is Estimated To Be Valued At US$ 1.29 Bn In 2022, And Is Forecast To Surpass US$ 2.2 Bn Valuation By The End Of 2032

Sales of surgical meshes are expected to account for more than 21 Mn units by 2032-end, owing to their increasing application in untapped markets, says a Fact.MR analyst.

Fact.MR A Market Research and Competitive Intelligence Provider: The global surgical mesh market is estimated to exceed a valuation of US$ 1.29 Bn in 2022, and expand at a significant CAGR of 5.5% by value over the assessment period (2022-2032).

The availability of surgical meshes in absorbable and non-absorbable forms has expanded their application for temporary as well as permanent reinforcement. In recent years, demand for surgical meshes has escalated in aiding breast reconstruction as they reduce the exposure risk of the implant. Increasing health literacy in North America and Europe will create ample opportunities for surgical mesh manufacturers over the coming years.

Sedentary lifestyle and increasing obesity among the population have resulted in several chronic health issues. The consequent weakening of the muscles extends space for organ prolapse and hernia. Putting these organs back in place by stitching the muscles together can result in muscle tearing and the recurrence of prolapse. However, reinforcing the weakened muscles with the help of a surgical mesh has shown to decrease recurrence and increase the longevity of the repair.

For more insights into the Market, Get A Sample of this Report!

https://www.factmr.com/connectus/sample?flag=S&rep_id=6632

Key Takeaways from Market Study

Winning Strategy

To attract new customers, market players are focusing on portfolio enhancement. Robust investments in R&D are driving product innovation for key market players. Meshes inhibiting the growth of bacterial films and preventing tissue adhesions are luring new consumers. Collaboration of manufacturers with scientific personnel and operating surgeons have enabled bespoke designing of meshes to best fit patients needs.

Manufacturers are also aiming for portfolio expansion through acquisition and partnerships. Partnering with companies that offer a well-aligned portfolio has significantly increased consumer penetration for key manufacturers. However, augmenting relations with local players and operating surgeons will be a key determinant of the products commercial success.

For Comprehensive Insights Ask An Analyst Here

https://www.factmr.com/connectus/sample?flag=AE&rep_id=6632

Scientific collaborations and robust R&D investments have also guided product innovation and became a common strategic approach adopted by leading surgical mesh manufacturing companies to upscale their market presence.

For instance:

Surgical Mesh Industry Research by Category

Surgical Mesh Market by Product Type:

Surgical Mesh Market by Nature:

Surgical Mesh Market by Surgical Access:

Surgical Mesh Market by Use Case:

Surgical Mesh Market by Raw Material:

Surgical Mesh Market by Region:

Get Customization on this Report for Specific Research Solutions

https://www.factmr.com/connectus/sample?flag=RC&rep_id=6632

More Valuable Insights on Offer

Fact.MR, in its new offering, presents an unbiased analysis of the global surgical mesh market, presenting historical market data (2017-2021) and forecast statistics for the period of 2022-2032.

The study reveals essential insights on the basis of product type (synthetic, biosynthetic, biologic, hybrid/composite), nature of mesh (absorbable, non-absorbable, partially absorbable), surgical access (open surgery, laparoscopic surgery), use case (hernia repair, pelvic floor disorder treatment, breast reconstruction, others), and raw material (polypropylene, polyethylene terephthalate, expanded polytetrafluoroethylene, polyglycolic acid, decellularized dermis/ECM, others), across seven major regions (North America, Latin America, Europe, East Asia, South Asia & ASEAN, Oceania, MEA).

Fact. MRs Domain Knowledge in Healthcare Division

Expert analysis, actionable insights, and strategic recommendations of the highly seasoned healthcare team at Fact.MR helps clients from across the globe with their unique business intelligence needs

With a repertoire of over thousand reports and 1 million-plus data points, the team has analysed the healthcare domain across 50+ countries for over a decade. The team provides unmatched end-to-end research and consulting services.

Check out more studies related to Healthcare Industry, conducted by Fact.MR:

Induced Pluripotent Stem Cell (iPSC) Market - Induced Pluripotent Stem Cell (iPSC) Market Analysis, By Cell Type (Vascular Cells, Cardiac Cells, Neuronal Cells, Liver Cells, Immune Cells), By Research Method (Cellular Reprogramming, Cell Culture, Cell Differentiation, Cell Analysis, Cellular Engineering), By Application (Drug Development & Toxicology Testing, Academic Research, Regenerative Medicine) - Global Market Insights 2022 to 2026

Newborn Screening Market -Newborn Screening Market by Product (Newborn Screening Instruments, Consumables), by Test Type (Dry Blood Spot Tests, Hearing Tests, Critical Congenital Heart Disease (CCHD) Screening Tests), by Technology (Immunoassays & Enzymatic Screening Tests, Tandem Mass Spectrometry, Molecular Assays, Hearing Screening Technologies, Pulse Oximetry, Others), by End User (Clinical Laboratories, Hospitals) and by Region- 2022 to 2032

Doxorubicin Market - Doxorubicin Market Analysis, By Formulation (Lyophilized Doxorubicin Powder, Doxorubicin Solution), By Cancer Type (Breast Cancer, Prostate Cancer, Ovarian Cancer, Lung Cancer, Bladder Cancer, Stomach Cancer, Leukemia), By Distribution Channel (Hospital Pharmacies, Retail Pharmacies, e-Commerce) - Global Market Insights 2022 to 2026

Microplate Systems Market - Microplate Systems Market By Product (Readers, Pipetting Systems & Dispensers, Washers), By End User (Biotechnology & Pharmaceuticals, Hospitals & Diagnostic Laboratories, Research Institutes, Academic Institutes) & Region - Global Market Insights 2022 to 2026

Drug Discovery Services Market - Drug Discovery Services Market Analysis by Process (Target Selection, Target Validation, Hit-To-Lead Identification, Lead Optimization and Candidate Validation), by Type (Medicinal Chemistry, Biology Services, Drug Metabolism and Pharmacokinetics) Region Forecast- 2022-2032

Lab Automation Market - Lab Automation Market by Product (Equipment, Microplate Readers, Software & Informatics, Automated ELISA Systems, Automated Nucleic Acid Purification Systems), by Application (Drug Discovery, Clinical Diagnostics, Genomics Solutions, Proteomics Solutions), & Region - Forecast to 2021-2031

Animal Model Market - Animal Model Market Analysis by Species (Rats, Mice, Guinea Pig, Rabbits, Monkeys, Dogs, Pigs, Cats, and Other Species), by Application (Basic & Applied Research and Drug Discovery/Development) and Region Forecast- 2022-2032

Blood Flow Measurement Devices Market - Blood Flow Measurement Devices Market Analysis by Product (Ultrasound - Doppler Ultrasound, Transit time Flow Meters (TTFM), Laser Doppler), Application (Non-invasive - Cardiovascular Disease, Diabetes, Dermatology, Invasive - CABG, Microvascular Surgery), by Region - Global Forecast 2022-2032

Contrast Media Injectors Market - Contrast Media Injectors Market by Product (Consumables, Injector Systems, Accessories), Application (Radiology, Interventional Radiology, Interventional Cardiology), End-User (Hospitals, Diagnostic Imaging Centers), & Regional Analysis till 2032

Preclinical Imaging Market - Preclinical Imaging Market Analysis By Product (Modality, and Reagents), and Region Forecast- 2022-2032

About Us:

Market research and consulting agency with a difference! Thats why 80% of Fortune 1,000 companies trust us for making their most critical decisions. While our experienced consultants employ the latest technologies to extract hard-to-find insights, we believe our USP is the trust clients have on our expertise. Spanning a wide range from automotive & industry 4.0 to healthcare & retail, our coverage is expansive, but we ensure even the most niche categories are analyzed. Our sales offices in United States and Dublin, Ireland. Headquarter based in Dubai, UAE. Reach out to us with your goals, and well be an able research partner.

Contact:

Mahendra Singh US Sales Office: 11140 Rockville Pike Suite 400 Rockville, MD 20852 Email: sales@factmr.com Tel: +1 (628) 251-158

Read the original post:
Technical Advancements & Innovative Products Likely to Expand Application of Surgical Meshes in Untapped Domains, States Fact.MR - BioSpace

Induced Pluripotent Stem Cells: Problems and Advantages when Applying …

Acta Naturae. 2010 Jul; 2(2): 1828.

Institute of Cytology and Genetics, Siberian Branch, Russian Academy of Sciences

Institute of Cytology and Genetics, Siberian Branch, Russian Academy of Sciences

Institute of Cytology and Genetics, Siberian Branch, Russian Academy of Sciences

Research Center of Clinical and Experimental Medicine, Siberian Branch, Russian Academy of Medical Sciences

Institute of Cytology and Genetics, Siberian Branch, Russian Academy of Sciences

Research Center of Clinical and Experimental Medicine, Siberian Branch, Russian Academy of Medical Sciences

This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Induced pluripotent stem cells (iPSCs) are a new type of pluripotent cells that can be obtained by reprogramming animal and human differentiated cells. In this review, issues related to the nature of iPSCs are discussed and different methods of iPSC production are described. We particularly focused on methods of iPSC production without the genetic modification of the cell genome and with means for increasing the iPSC production efficiency. The possibility and issues related to the safety of iPSC use in cell replacement therapy of human diseases and a study of new medicines are considered.

Keywords: induced pluripotent stem cells, directed stem cell differentiation, cell replacement therapy

Pluripotent stem cells are a unique model for studying a variety of processes that occur in the early development of mammals and a promising tool in cell therapy of human diseases. The unique nature of these cells lies in their capability, when cultured, for unlimited selfrenewal and reproduction of all adult cell types in the course of their differentiation [1]. Pluripotency is supported by a complex system of signaling molecules and gene network that is specific for pluripotent cells. The pivotal position in the hierarchy of genes implicated in the maintenance of pluripotency is occupied by Oct4, Sox2 , and Nanog genes encoding transcription factors [2, 3]. The mutual effect of outer signaling molecules and inner factors leads to the formation of a specific expression pattern, as well as to the epigenome state characteristic of stem cells. Both spontaneous and directed differentiations are associated with changes in the expression pattern and massive epigenetic transformations, leading to transcriptome and epigenome adjustment to a distinct cell type.

Until recently, embryonic stem cells (ESCs) were the only wellstudied source of pluripotent stem cells. ESCs are obtained from either the inner cell mass or epiblast of blastocysts [46]. A series of protocols has been developed for the preparation of various cell derivatives from human ESCs. However, there are constraints for ESC use in cell replacement therapy. The first constraint is the immune incompatibility between the donor cells and the recipient, which can result in the rejection of transplanted cells. The second constraint is ethical, because the embryo dies during the isolation of ESCs. The first problem can be solved by the somatic cell nuclear transfer into the egg cell and then obtaining the embryo and ESCs. The nuclear transfer leads to genome reprogramming, in which ovarian cytoplasmic factors are implicated. This way of preparing pluripotent cells from certain individuals was called therapeutic cloning. However, this method is technologyintensive, and the reprogramming yield is very low. Moreover, this approach encounters the abovementioned ethic problem that, in this case, is associated with the generation of many human ovarian cells [7].

In 2006, the preparation of pluripotent cells by the ectopic expression of four genes Oct4 , Sox2 , Klf4 , and cMyc in both embryonic and adult murine fibroblasts was first reported [8]. The pluripotent cells derived from somatic ones were called induced pluripotent stem cells (iPSCs). Using this set of factors (Oct4, Sox2, Klf4, and cMyc), iPSCs were prepared later from various differentiated mouse [914] and human [1517] cell types. Human iPSCs were obtained with a somewhat altered gene set: Oct4 , Sox2 , Nanog , and Lin28 [18]. Induced PSCs closely resemble ESCs in a broad spectrum of features. They possess similar morphologies and growth manners and are equally sensitive to growth factors and signaling molecules. Like ESCs, iPSCs can differentiate in vitro into derivatives of all three primary germ layers (ectoderm, mesoderm, and endoderm) and form teratomas following their subcutaneous injection into immunodeficient mice. Murine iPSCs injected into blastocysts are normally included in the development to yield animals with a high degree of chimerism. Moreover, murine iPSCs, when injected into tetraploid blastocycts, can develop into a whole organism [19, 20]. Thus, an excellent method that allows the preparation of pluripotent stem cells from various somatic cell types while bypassing ethical problems has been uncovered by researchers.

In the first works on murine and human iPSC production, either retro or lentiviral vectors were used for the delivery of Oct4 , Sox2 , Klf4 , and cMyc genes into somatic cells. The efficiency of transduction with retroviruses is high enough, although it is not the same for different cell types. Retroviral integration into the host genome requires a comparatively high division rate, which is characteristic of the relatively narrow spectrum of cultured cells. Moreover, the transcription of retroviral construct under the control of a promoter localized in 5LTR (long terminal repeat) is terminated when the somatic cell transform switches to the pluripotent state [21]. This feature makes retroviruses attractive in iPSC production. Nevertheless, retroviruses possess some properties that make iPSCs that are produced using them improper for cell therapy of human diseases. First, retroviral DNA is integrated into the host cell genome. The integration occurs randomly; i.e., there are no specific sequences or apparent logic for retroviral integration. The copy number of the exogenous retroviral DNA that is integrated into a genome may vary to a great extent [15]. Retroviruses being integrated into the cell genome can introduce promoter elements and polyadenylation signals; they can also interpose coding sequences, thus affecting transcription. Second, since the transcription level of exogenous Oct4 , Sox2 , Klf4 , and cMyc in the retroviral construct decreases with cell transition into the pluripotent state, this can result in a decrease in the efficiency of the stable iPSC line production, because the switch from the exogenous expression of pluripotency genes to their endogenous expression may not occur. Third, some studies show that the transcription of transgenes can resume in the cells derived from iPSCs [22]. The high probability that the ectopic Oct4 , Sox2 , Klf4 , and cMyc gene expression will resume makes it impossible to apply iPSCs produced with the use of retroviruses in clinical trials; moreover, these iPSCs are hardly applicable even for fundamental studies on reprogramming and pluripotency principles. Lentiviruses used for iPSC production can also be integrated into the genome and maintain their transcriptional activity in pluripotent cells. One way to avoid this situation is to use promoters controlled by exogenous substances added to the culture medium, such as tetracycline and doxycycline, which allows the transgene transcription to be regulated. iPSCs are already being produced using such systems [23].

Another serious problem is the gene set itself that is used for the induction of pluripotency [22]. The ectopic transcription of Oct4 , Sox2 , Klf4 , and cMyc can lead to neoplastic development from cells derived from iPSCs, because the expression of Oct4 , Sox2 , Klf4, and cMyc genes is associated with the development of multiple tumors known in oncogenetics [22, 24]. In particular, the overexpression of Oct4 causes murine epithelial cell dysplasia [25], the aberrant expression of Sox2 causes the development of serrated polyps and mucinous colon carcinomas [26], breast tumors are characterized by elevated expression of Klf4 [27] , and the improper expression of cMyc is observed in 70% of human cancers [28]. Tumor development is oberved in ~50% of murine chimeras obtained through the injection of retroviral iPSCs into blastocysts, which is very likely associated with the reactivation of exogenous cMyc [29, 30].

Several possible strategies exist for resolving the above-mentioned problems:

The search for a less carcinogenic gene set that is necessary and sufficient for reprogramming;

The minimization of the number of genes required for reprogramming and searching for the nongenetic factors facilitating it;

The search for systems allowing the elimination of the exogenous DNA from the host cell genome after the reprogramming;

The development of delivery protocols for nonintegrated genetic constructs;

The search for ways to reprogram somatic cells using recombinant proteins.

The ectopic expression of cMyc and Klf4 genes is the most dangerous because of the high probability that malignant tumors will develop [22]. Hence the necessity to find other genes that could substitute cMyc and Klf4 in iPSC production. It has been reported that these genes can be successfully substituted by Nanog and Lin28 for reprogramming human somatic cells [18;] . iPSCs were prepared from murine embryonic fibroblasts by the overexpression of Oct4 and Sox2 , as well as the Esrrb gene encoding the murine orphan nuclear receptor beta. It has already been shown that Esrrb , which acts as a transcription activator of Oct4 , Sox2 , and Nanog , is necessary for the selfrenewal and maintenance of the pluripotency of murine ESCs. Moreover, Esrrb can exert a positive control over Klf4 . Thus, the genes causing elevated carcinogenicity of both iPSCs and their derivatives can be successfully replaced with less dangerous ones [31].

The Most Effectively Reprogrammed Cell Lines . Murine and human iPSCs can be obtained from fibroblasts using the factors Oct4, Sox2, and Klf4, but without cMyc . However, in this case, reprogramming decelerates and an essential shortcoming of stable iPSC clones is observed [32, 33]. The reduction of a number of necessary factors without any decrease in efficiency is possible when iPSCs are produced from murine and human neural stem cells (NSCs) [12, 34, 35]. For instance, iPSCs were produced from NSCs isolated from adult murine brain using two factors, Oct4 and Klf4, as well as even Oct4 by itself [12, 34]. Later, human iPSCs were produced by the reprogramming of fetal NSCs transduced with a retroviral vector only carrying Oct4 [35] . It is most likely that the irrelevance of Sox2, Klf4, and cMyc is due to the high endogenous expression level of these genes in NSCs.

Successful reprogramming was also achieved in experiments with other cell lines, in particular, melanocytes of neuroectodermal genesis [36]. Both murine and human melanocytes are characterized by a considerable expression level of the Sox2 gene, especially at early passages. iPSCs from murine and human melanocytes were produced without the use of Sox2 or cMyc. However, the yield of iPSC clones produced from murine melanocytes was lower (0.03% without Sox2 and 0.02% without cMyc) in comparison with that achieved when all four factors were applied to melanocytes (0.19%) and fibroblasts (0.056%). A decreased efficiency without Sox2 or cMyc was observed in human melanocyte reprogramming (0.05% with all four factors and 0.01% without either Sox2 or cMyc ). All attempts to obtain stable iPSC clones in the absence of both Sox2 and cMyc were unsuccessful [36]. Thus, the minimization of the number of factors required for iPSC preparation can be achieved by choosing the proper somatic cell type that most effectively undergoes reprogramming under the action of fewer factors, for example, due to the endogenous expression of pluripotency genes. However, if human iPSCs are necessary, these somatic cells should be easily accessible and wellcultured and their method of isolation should be as noninvasive as possible.

One of these cell types can be adipose stem cells (ASCs). This is a heterogeneous group of multipotent cells which can be relatively easily isolated in large amounts from adipose tissue following liposuction. Human iPSCs were successfully produced from ASCs with a twofold reprogramming rate and 20fold efficiency (0.2%), exceeding those of fibroblasts [37].

However, more accessible resources for the effective production of human iPSCs are keratinocytes. When compared with fibroblasts, human iPSC production from keratinocytes demonstrated a 100fold greater efficiency and a twofold higher reprogramming rate [38].

It has recently been found that the reprogramming of murine papillary dermal fibroblasts (PDFs) into iPSCs can be highly effective with the overexpression of only two genes, Oct4 and Klf4 , inserted into retroviral vectors [39;]. PDFs are specialized cells of mesodermal genesis surrounding the stem cells of hair follicles . One characteristic feature of these cells is the endogenous expression of Sox2 , Klf4 , and cMyc genes, as well as the geneencoding alkaline phosphatase, one of the murine and human ESC markers. PDFs can be easily separated from other cell types by FACS (fluorescenceactivated cell sorting) using life staining with antibodies against the surface antigens characteristic of one or another cell type. The PDF reprogramming efficiency with the use of four factors (Oct4, Sox2, Klf4, and cMyc) retroviral vectors is 1.38%, which is 1,000fold higher than the skin fibroblast reprogramming efficiency in the same system. Reprogramming PDFs with two factors, Oct4 and Klf4 , yields 0.024%, which is comparable to the efficiency of skin fibroblast reprogramming using all four factors. The efficiency of PDF reprogramming is comparable with that of NSCs, but PDF isolation is steady and far less invasive [39]. It seems likely that human PDF lines are also usable, and this cell type may appear to be one of the most promising for human iPSC production in terms of pharmacological studies and cell replacement therapy. The use of such cell types undergoing more effective reprogramming, together with methods providing the delivery of pluripotency genes without the integration of foreign DNA into the host genome and chemical compounds increasing the reprogramming efficiency and substituting some factors required for reprogramming, is particularly relevant.

Chemical Compounds Increasing Cell Reprogramming Efficiency. As was noted above, the minimization of the factors used for reprogramming decreases the efficiency of iPSC production. Nonetheless, several recent studies have shown that the use of genetic mechanisms, namely, the initiation of ectopic gene expression, can be substituted by chemical compounds, most of them operating at the epigenetic level. For instance, BIX01294 inhibiting histone methyltransferase G9a allows murine fibroblast reprogramming using only two factors, Oct4 and Klf4, with a fivefold increased yield of iPSC clones in comparison with the control experiment without BIX01294 [40]. BIX01294 taken in combination with another compound can increase the reprogramming efficiency even more. In particular, BIX01294 plus BayK8644 elevated the yield of iPCSs 15 times, and BIX01294 plus RG108 elevated it 30 times when only two reprogramming factors, Oct4 and Klf4, were used. RG108 is an inhibitor of DNA methyltransferases, and its role in reprogramming is apparently in initiating the more rapid and effective demethylation of promoters of pluripotent cellspecific genes, whereas BayK8644 is an antagonist of Ltype calcium channels, and its role in reprogramming is not understood very well [40]. However, more considerable results were obtained in reprogramming murine NSCs. The use of BIX01294 allowed a 1.5fold increase in iPSC production efficiency with two factors, Oct4 and Klf4, in comparison with reprogramming with all four factors. Moreover, BIX01294 can even substitute Oct4 in the reprogramming of NSCs, although the yield is very low [41]. Valproic (2propylvaleric) acid inhibiting histone deacetylases can also substitute cMyc in reprogramming murine and human fibroblasts. Valproic acid (VPA) increases the reprogramming efficiency of murine fibroblasts 50 times, and human fibroblasts increases it 1020 times when three factors are used [42, 43]. Other deacetylase inhibitors, such as TSA (trichostatin A) and SAHA (suberoylanilide hyroxamic acid), also increase the reprogramming efficiency. TSA increases the murine fibroblast reprogramming efficiency 15 times, and SAHA doubles it when all four factors are used [42]. Besides epigenetic regulators, the substances inhibiting the protein components of signaling pathways implicated in the differentiation of pluripotent cells are also applicable in the substitution of reprogramming factors. In particular, inhibitors of MEK and GSK3 kinases (PD0325901 and CHIR99021, respectively) benefit the establishment of the complete and stable pluripotency of iPSCs produced from murine NSCs using two factors, Oct4 and Klf4 [41, 44].

It has recently been shown that antioxidants can considerably increase the efficiency of somatic cell reprogramming. Ascorbic acid (vitamin C) can essentially influence the efficiency of iPSC production from various murine and human somatic cell types [45]. The transduction of murine embryonic fibroblasts (mEFs) with retroviruses carrying the Oct4 , Sox2 , and Klf4 genes results in a significant increase in the production level of reactive oxygen species (ROS) compared with that of both control and Efs tranduced with Oct4 , Sox2 , cMyc , and Klf4 . In turn, the increase in the ROS level causes accelerated aging and apoptosis of the cell, which should influence the efficiency of cell reprogramming. By testing several substances possessing antioxidant activity such as vitamin B1, sodium selenite, reduced glutathione, and ascorbic acid, the authors have found that combining these substances increases the yield of GFPpositive cells in EF reprogramming (the Gfp gene was under the control of the Oct4 gene promoter). The use of individual substances has shown that only ascorbate possesses a pronounced capability to increase the level of GFPpositive cells, although other substances keep their ROSdecreasing ability. In all likelihood, this feature of ascorbates is not directly associated with its antioxidant activity [45]. The score of GFPpositive iPSC colonies expressing an alkaline phosphatase has shown that the efficiency of iPSC production from mEFs with three factors (Oct4, Sox2, and Klf4) can reach 3.8% in the presence of ascorbate. When all four factors (Oct4, Sox2, Klf4, and cMyc) are used together with ascorbate, the efficiency of iPSC production may reach 8.75%. A similar increase in the iPSC yield was also observed in the reprogramming of murine breast fibroblasts; i.e., the effect of vitamin C is not limited by one cell type. Moreover, the effect of vitamin C on the reprogramming efficiency is more profound than that of the deacetylase inhibitor valproic (2propylvaleric) acid. The mutual effect of ascorbate and valproate is additive; i.e., these substances have different action mechanisms. Moreover, vitamin C facilitates the transition from preiPSCs to stable pluripotent cells. This feature is akin to the effects of PD0325901 and CHIR99021, which are inhibitors of MEK and GSK3 kinases, respectively. This effect of vitamin C expands to human cells as well [45]. Following the transduction of human fibroblasts with retroviruses carrying Oct4 , Sox2 , Klf4 , and cMyc and treatment with ascorbate, the authors prepared iPSCs with efficiencies reaching 6.2%. The reprogramming efficiency of ASCs under the same conditions reached 7.06%. The mechanism of the effect that vitamin C has on the reprogramming efficiency is not known in detail. Nevertheless, the acceleration of cell proliferation was observed at the transitional stage of reprogramming. The levels of the p53 and p21 proteins decreased in cells treated with ascorbate, whereas the DNA repair machinery worked properly [45]. It is interesting that an essential decrease in the efficiency of iPSC production has been shown under the action of processes initiated by p53 and p21 [4650].

As was mentioned above, for murine and human iPSC production, both retro and lentiviruses were initially used as delivery vectors for the genes required for cell reprogramming. The main drawback of this method is the uncontrolled integration of viral DNA into the host cells genome. Several research groups have introduced methods for delivering pluripotency genes into the recipient cell which either do not integrate allogenic DNA into the host genome or eliminate exogenous genetic constructs from the genome.

CreloxP Mediated Recombination. To prepare iPSCs from patients with Parkinsons disease, lentiviruses were used, the proviruses of which can be removed from the genome by Cre recombinase. To do this, the loxP site was introduced into the lentiviral 3LTRregions containing separate reprogramming genes under the control of the doxycyclineinducible promoter. During viral replication, loxP was duplicated in the 5LTR of the vector. As a result, the provirus integrated into the genome was flanked with two loxP sites. The inserts were eliminated using the temporary transfection of iPSCs with a vector expressing Cre recombinase [51].

In another study, murine iPSCs were produced using a plasmid carrying the Oct4 , Sox2 , Klf4I, and cMyc genes in the same reading frame in which individual cDNAs were separated by sequences encoding 2 peptides, and practically the whole construct was flanked with loxP sites [52]. The use of this vector allowed a notable decrease in the number of exogenous DNA inserts in the host cells genome and, hence, the simplification of their following excision [52]. It has been shown using lentiviruses carrying similar polycistronic constructs that one copy of transgene providing a high expression level of the exogenous factors Oct4, Sox2, Klf4, and cMyc is sufficient for the reprogramming of differentiated cells into the pluripotent state [53, 54].

The drawback of the CreloxP system is the incomplete excision of integrated sequences; at least the loxP site remains in the genome, so the risk of insertion mutations remains.

Plasmid Vectors . The application of lentiviruses and plasmids carrying the loxP sites required for the elimination of transgene constructs modifies, although insignificantly, the host cells genome. One way to avoid this is to use vector systems that generally do not provide for the integration of the whole vector or parts of it into the cells genome. One such system providing a temporary transfection with polycistronic plasmid vectors was used for iPSC production from mEFs [29]. A polycistronic plasmid carrying the Oct4 , Sox2 , and Klf4 gene cDNAs, as well as a plasmid expressing cMyc , was transfected into mEFs one, three, five, and seven days after their primary seeding. Fibroblasts were passaged on the ninth day, and the iPSC colonies were selected on the 25th day. Seven out of ten experiments succeeded in producing GFPpositive colonies (the Gfp gene was under the control of the Nanog gene promoter). The iPSCs that were obtained were similar in their features to murine ESCs and did not contain inserts of the used DNA constructs in their genomes. Therefore, it was shown that wholesome murine iPSCs that do not carry transgenes can be reproducibly produced, and that the temporary overexpression of Oct4 , Sox2 , Klf4 , and cMyc is sufficient for reprogramming. The main drawback of this method is its low yield. In ten experiments the yield varied from 1 to 29 iPSC colonies per ten million fibroblasts, whereas up to 1,000 colonies per ten millions were obtained in the same study using retroviral constructs [29].

Episomal Vectors . Human iPSCs were successfully produced from skin fibroblasts using single transfection with polycistronic episomal constructs carrying various combinations of Oct4 , Sox2 , Nanog , Klf4 , cMyc , Lin28 , and SV40LT genes. These constructs were designed on the basis of the oriP/EBNA1 (EpsteinBarr nuclear antigen1) vector [55]. The oriP/EBNA1 vector contains the IRES2 linker sequence allowing the expression of several individual cDNAs (encoding the genes required for successful reprogramming in this case) into one polycistronic mRNA from which several proteins are translated. The oriP/EBNA1 vector is also characterized by lowcopy representation in the cells of primates and can be replicated once per cell cycle (hence, it is not rapidly eliminated, the way common plasmids are). Under nonselective conditions, the plasmid is eliminated at a rate of about 5% per cell cycle [56]. In this work, the broad spectrum of the reprogramming factor combinations was tested, resulting in the best reprogramming efficiency with cotransfection with three episomes containing the following gene sets: Oct4 + Sox2 + Nanog + Klf4 , Oct4 + Sox2 + SV40LT + Klf4 , and cMyc + Lin28 . SV40LT ( SV40 large T gene ) neutralizes the possible toxic effect of overexpression [57]. The authors have shown that wholesome iPSCs possessing all features of pluripotent cells can be produced following the temporary expression of a certain gene combination in human somatic cells without the integration of episomal DNA into the genome. However, as in the case when plasmid vectors are being used, this way of reprogramming is characterized by low efficiency. In separate experiments the authors obtained from 3 to 6 stable iPSC colonies per 106 transfected fibroblasts [55]. Despite the fact that skin fibroblasts are wellcultured and accessible, the search for other cell types which are relatively better cultured and more effectively subject themselves to reprogramming through this method is very likely required. Another drawback of the given system is that this type of episome is unequally maintained in different cell types.

PiggyBacTransposition . One promising system used for iPSC production without any modification of the host genome is based on DNA transposons. Socalled PiggyBac transposons containing 2linkered reprogramming genes localized between the 5 and 3terminal repeats were used for iPSC production from fibroblasts. The integration of the given constructs into the genome occurs due to mutual transfection with a plasmid encoding transposase. Following reprogramming due to the temporary expression of transposase, the elimination of inserts from the genome took place [58, 59]. One advantage of the PiggyBac system on CreloxP is that the exogenous DNA is completely removed [60].

However, despite the relatively high efficiency of exogenous DNA excision from the genome by PiggyBac transposition, the removal of a large number of transposon copies is hardly achievable.

Nonintegrating Viral Vectors . Murine iPSCs were successfully produced from hepatocytes and fibroblasts using four adenoviral vectors nonintegrating into the genome and carrying the Oct4 , Sox2 , Klf4 , and cMyc genes. An analysis of the obtained iPSCs has shown that they are similar to murine ESCs in their properties (teratoma formation, gene promoter DNA methylation, and the expression of pluripotent markers), but they do not carry insertions of viral DNA in their genomes [61]. Later, human fibroblastderived iPSCs were produced using this method [62].

The authors of this paper cited the postulate that the use of adenoviral vectors allows the production of iPSCs, which are suitable for use without the risk of viral or oncogenic activity. Its very low yield (0.00010.001%), the deceleration of reprogramming, and the probability of tetraploid cell formation are the drawbacks of the method. Not all cell types are equally sensitive to transduction with adenoviruses.

Another method of gene delivery based on viral vectors was recently employed for the production of human iPSCs. The sendaivirus (SeV)based vector was used in this case [63]. SeV is a singlestranded RNA virus which does not modify the genome of recipient cells; it seems to be a good vector for the expression of reprogramming factors. Vectors containing either all pluripotency factors or three of them (without ) were used for reprogramming the human fibroblast. The construct based on SeV is eliminated later in the course of cell proliferation. It is possible to remove cells with the integrated provirus via negative selection against the surface HN antigen exposed on the infected cells. The authors postulate that reprogramming technology based on SeV will enable the production of clinically applicable human iPSCs [63].

Cell Transduction with Recombinant Proteins . Although the methods for iPSC production without gene modification of the cells genome (adenoviral vectors, plasmid gene transfer, etc.) are elaborated, the theoretical possibility for exogenous DNA integration into the host cells genome still exists. The mutagenic potential of the substances used presently for enhancing iPSC production efficiency has not been studied in detail. Fully checking iPSC genomes for exogenous DNA inserts and other mutations is a difficult task, which becomes impossible to solve in bulk culturing of multiple lines. The use of protein factors delivered into a differentiated cell instead of exogenous DNA may solve this problem. Two reports have been published to date in which murine and human iPSCs were produced using the recombinant Oct4, Sox2, Klf4, and cMyc proteins [64, 65] . T he method used to deliver the protein into the cell is based on the ability of peptides enriched with basic residues (such as arginine and lysine) to penetrate the cells membrane. Murine iPSCs were produced using the recombinant Oct4, Sox2, Klf4, and cMyc proteins containing eleven Cterminal arginine residues and expressed in E. coli . The authors succeeded in producing murine iPSCs during four rounds of protein transduction into embryonic fibroblasts [65]. However, iPSCs were only produced when the cells were additionally treated with 2propylvalerate (the deacetylase inhibitor). The same principle was used for the production of human iPSCs, but protein expression was carried out in human HEK293 cells, and the proteins were expressed with a fragment of nine arginins at the protein Cend. Researchers have succeeded in producing human iPSCs after six transduction rounds without any additional treatment [64]. The efficiency of producing human iPSC in this way was 0.001%, which is one order lower than the reprogramming efficiency with retroviruses. Despite some drawbacks, this method is very promising for the production of patientspecific iPSCs.

The first lines of human pluripotent ESCs were produced in 1998 [6]. In line with the obvious fundamental importance of embryonic stem cell studies with regard to the multiple processes taking place in early embryogenesis, much of the interest of investigators is associated with the possibility of using ESCs and their derivatives as models for the pathogenesis of human diseases, new drugs testing, and cell replacement therapy. Substantial progress is being achieved in studies on directed human ESC differentiation and the possibility of using them to correct degenerative disorders. Functional cell types, such as motor dopaminergic neurons, cardiomyocytes, and hematopoietic cell progenitors, can be produced as a result of ESC differentiation. These cell derivatives, judging from their biochemical and physiological properties, are potentially applicable for the therapy of cardiovascular disorders, nervous system diseases, and human hematological disorders [66]. Moreover, derivatives produced from ESCs have been successfully used for treating diseases modeled on animals. Therefore, bloodcell progenitors produced from ESCs were successfully used for correcting immune deficiency in mice. Visual functions were restored in blind mice using photoreceptors produced from human ESCs, and the normal functioning of the nervous system was restored in rats modeling Parkinsons disease using the dopaminergic neurons produced from human ESCs [6770]. Despite obvious success, the fullscale application of ESCs in therapy and the modeling of disorders still carry difficulties, because of the necessity to create ESC banks corresponding to all HLAhaplotypes, which is practically unrealistic and hindered by technical and ethical problems.

Induced pluripotent stem cells can become an alternative for ESCs in the area of clinical application of cell replacement therapy and screening for new pharmaceuticals. iPSCs closely resemble ESCs and, at the same time, can be produced in almost unlimited amounts from the differentiated cells of each patient. Despite the fact that the first iPSCs were produced relatively recently, work on directed iPSC differentiation and the production of patientspecific iPSCs is intensive, and progress in this field is obvious.

Dopamine and motor neurons were produced from human iPSCs by directed differentiation in vitro [71, 72]. These types of neurons are damaged in many inherited or acquired human diseases, such as spinal cord injury, Parkinsons disease, spinal muscular atrophy, and amyotrophic lateral sclerosis. Some investigators have succeeded in producing various retinal cells from murine and human iPSCs [7375]. Human iPSCs have been shown to be spontaneously differentiated in vitro into the cells of retinal pigment epithelium [76]. Another group of investigators has demonstrated that treating human and murine iPSCs with Wnt and Nodal antagonists in a suspended culture induces the appearance of markers of cell progenitors and pigment epithelium cells. Further treating the cells with retinoic acid and taurine activates the appearance of cells expressing photoreceptor markers [75].

Several research groups have produced functional cardiomyocytes (CMs) in vitro from murine and human iPSCs [7781]. Cardiomyocytes produced from iPSC are very similar in characteristics (morphology, marker expression, electrophysiological features, and sensitivity to chemicals) to the CMs of cardiac muscle and to CMs produced from differentiated ESCs. Moreover, murine iPSCs, when injected, can repair muscle and endothelial cardiac tissues damaged by cardiac infarction [77].

Hepatocytelike cell derivatives, dendritic cells, macrophages, insulinproducing cell clusters similar to the duodenal islets of Langerhans, and hematopoietic and endothelial cells are currently produced from murine and human iPSCs, in addition to the alreadylisted types of differentiated cells [8285].

In addition to directed differentiation in vitro , investigators apply much effort at producing patientspecific iPSCs. The availability of pluripotent cells from individual patients makes it possible to study pathogenesis and carry out experiments on the therapy of inherited diseases, the development of which is associated with distinct cell types that are hard to obtain by biopsy: so the use of iPSCs provides almost an unlimited resource for these investigations. Recently, the possibility of treating diseases using iPSCs was successfully demonstrated, and the design of the experiment is presented in the figure. A mutant allele was substituted with a normal allele via homologous recombination in murine fibroblasts representing a model of human sickle cell anemia. iPSCs were produced from repaired fibroblasts and then differentiated into hematopoietic cell precursors. The hematopoietic precursors were then injected into a mouse from which the skin fibroblasts were initially isolated (). As a result, the initial pathological phenotype was substantially corrected [86]. A similar approach was applied to the fibroblasts and keratinocytes of a patient with Fanconis anemia. The normal allele of the mutant gene producing anemia was introduced into a somatic cell genome using a lentivirus, and then iPSCs were obtained from these cells. iPSCs carrying the normal allele were differentiated into hematopoietic cells maintaining a normal phenotype [87]. The use of lentiviruses is unambiguously impossible when producing cells to be introduced into the human body due to their oncogenic potential. However, new relatively safe methods of genome manipulation are currently being developed; for instance, the use of synthetic nucleases containing zinc finger domains allowing the effective correction of genetic defects in vitro [88].

Design of an experiment on repairing the mutant phenotype in mice modeling sickle cell anemia development [2]. Fibroblasts isolated from the tail of a mouse (1) carrying a mutant allele of the gene encoding the human hemoglobin -chain (hs) were used for iPSC production (2). The mutation was then repaired in iPSCs by means of homological recombination (3) followed by cell differentiation via the embryoid body formation (4). The directed differentiation of the embryoid body cells led to hematopoietic precursor cells (5) that were subsequently introduced into a mouse exposed to ionizing radiation (6).

The induced pluripotent stem cells are an excellent model for pathogenetic studies at the cell level and testing compounds possessing a possible therapeutic effect.

The induced pluripotent stem cells were produced from the fibroblasts of a patient with spinal muscular atrophy (SMA) (SMAiPSCs). SMA is an autosomal recessive disease caused by a mutation in the SMN1 ( survival motor neuron 1 ) gene, which is manifested as the selective nonviability of lower motor neurons. Patients with this disorder usually die at the age of about two years. Existing experimental models of this disorder based on the use of flatworms, drosophila, and mice are not satisfactory. The available fibroblast lines from patients with SMA cannot provide the necessary data on the pathogenesis of this disorder either. It was shown that motor neurons produced from SMAiPSCs can retain the features of SMA development, selective neuronal death, and the lack of SMN1 transcription. Moreover, the authors succeeded in elevating the SMN protein level and aggregation (encoded by the SMN2 gene, whose expression can compensate for the shortage in the SMN1 protein) in response to the treatment of motor neurons and astrocytes produced from SMAiPSCs with valproate and torbomycin [89;]. iPSCs and their derivatives can serve as objects for pharmacological studies, as has been demonstrated on iPSCs from patients with familial dysautonomia (FDA) [90]. FDA is an inherited autosomal recessive disorder manifested as the degeneration of sensor and autonomous neurons. This is due to a mutation causing the tissuespecific splicing of the IKBKAP gene, resulting in a decrease in the level of the fulllength IKAP protein. iPSCs were produced from fibroblasts of patients with FDA. They possessed all features of pluripotent cells. Neural derivatives produced from these cells had signs of FDA pathogenesis and low levels of the fulllength IKBKAP transcript. The authors studied the effect of three substances, kinetin, epigallocatechin gallate, and tocotrienol, on the parameters associated with FDA pathogenesis. Only kinetin has been shown to induce an increase in the level of fulllength IKBKAP transcript. Prolonged treatment with kinetin induces an increase in the level of neuronal differentiation and expression of peripheral neuronal markers.

Currently, a broad spectrum of iPSCs is produced from patients with various inherited pathologies and multifactorial disorders, such as Parkinsons disease, Down syndrome, type 1 diabetes, Duchenne muscular dystrophy, talassemia, etc., which are often lethal and can scarcely be treated with routine therapy [51, 87, 89, 9194]. The data on iPSCs produced by reprogramming somatic cells from patients with various pathologies are given in the .

Functional categories of M. tuberculosis genes with changed expression level during transition to the NC state

One can confidently state that both iPSCs themselves and their derivatives are potent instruments applicable in biomedicine, cell replacement therapy, pharmacology, and toxicology. However, the safe application of iPSCbased technologies requires the use of methods of iPSCs production and their directed differentiation which minimize both the possibility of mutations in cell genomes under in vitro culturing and the probability of malignant transformation of the injected cells. The development of methods for human iPSC culturing without the use of animal cells (for instance, the feeder layer of murine fibroblasts) is necessary; they make a viralorigin pathogen transfer from animals to humans impossible. There is a need for the maximum standardization of conditions for cell culturing and differentiation.

This study was supported by the Russian Academy of Sciences Presidium Program Molecular and Cell Biology.

The rest is here:
Induced Pluripotent Stem Cells: Problems and Advantages when Applying ...

Umoja Biopharma Presents Data on its Engineered Induced Pluripotent Stem Cell Platform at the 2022 International Society for Stem Cell Research Annual…

SEATTLE, June 16, 2022 (GLOBE NEWSWIRE) -- Umoja Biopharma, Inc., an immuno-oncology company pioneering off-the-shelf, integrated therapeutics that reprogram immune cells in vivo to treat patients with solid and hematologic malignancies, announced today that it will have a poster presentation at the 2022 International Society for Stem Cell Research (ISSCR) Annual Meeting, to be held June 15-18, 2022 in San Francisco, California.

On Wednesday, June 15th, Principal Scientist & iPSC Team Lead, Teisha Rowland, Ph.D., will give a poster presentation titled, A Synthetic Cytokine Receptor Platform for Producing Cytotoxic Innate Lymphocytes as Off-the-Shelf Cancer Therapeutics. The presentation will discuss Umojas engineered induced pluripotent stem cell (iPSC) platform, that incorporates the synthetic cytokine receptor system rapamycin-activated cytokine receptor (RACR) platform. Umojas engineered iPSCs that are modified to express RACR, called RACR-induced cytotoxic innate lymphoid (iCIL) cells, drive differentiation and expansion of the cells while eliminating the need for expensive cytokines and other raw materials. The RACR platform has the potential to enable cytokine-free manufacturing and engraftment of the engineered cells in the patient without the need for toxic lymphodepletion.

Despite the advances chimeric antigen receptor T cell therapies have provided to the oncology space, we continue to battle significant challenges that these therapies cannot address, like limited expansion capacity and scalability, manufacturing complexity, variability among patients, and the need for toxic chemotherapy administration to combat patients anti-allograft response, said Andy Scharenberg, M.D., co-founder and Chief Executive Officer of Umoja. We are developing an engineered iPSC platform, including the RACR platform, to address these challenges by enabling a scalable, virtually unlimited, and simplified manufacturing of engineered, cancer-fighting cytotoxic innate lymphocytes.

About Umoja Biopharma

Umoja Biopharma, Inc. is an early clinical-stage company advancing an entirely new approach to immunotherapy. Umoja Biopharma, Inc. is a transformative multi-platform immuno-oncology company founded with the goal of creating curative treatments for solid and hematological malignancies by reprogramming immune cells in vivo to target and fight cancer. Founded based on pioneering work performed at Seattle Childrens Research Institute and Purdue University, Umojas novel approach is powered by integrated cellular immunotherapy technologies including the VivoVec in vivo delivery platform, the RACR/CAR in vivo cell expansion/control platform, and the TumorTag targeting platform. Designed from the ground up to work together, these platforms are being developed to create and harness a powerful immune response in the body to directly, safely, and controllably attack cancer. Umoja believes that its approach can provide broader access to the most advanced immunotherapies and enable more patients to live better, fuller lives. To learn more, visit http://umoja-biopharma.com/.

About RACR

CAR T cells generated by the body with VivoVec can be expanded and sustained with the rapamycin activated cytokine receptor (RACR) system, an engineered signaling system designed to improve chimeric antigen receptor (CAR) T cell persistence and produce durable anti-tumor responses. The RACR/CAR payload is integrated into the genomic DNA of a patients T cells. Rapamycin activates the RACR system resulting in preferential expansion and survival of cancer-fighting T cells. The RACR technology enables a patients cells to expand in a manner that resembles a natural immune response that does not require lymphodepletion, promoting durable T cell engraftment. RACR/CAR technology can also be used to enhanceex vivomanufacturing in support of more traditional autologous or allogeneic cell therapy manufacturing processes. To learn more about Umojas RACR platform please visit https://www.umoja-biopharma.com/platforms/

Media Contact: Darren Opland, Ph.D. LifeSci Communications darren@lifescicomms.com

Link:
Umoja Biopharma Presents Data on its Engineered Induced Pluripotent Stem Cell Platform at the 2022 International Society for Stem Cell Research Annual...

Induced Pluripotent Stem Cell (iPSC)Derived Lymphocytes for Adoptive …

Curr Hematol Malig Rep. 2019; 14(4): 261268.

Department of Hematology, Amsterdam University Medical Centers, Cancer Center Amsterdam, Location VUmc, Amsterdam, Netherlands

Department of Hematology, Amsterdam University Medical Centers, Cancer Center Amsterdam, Location VUmc, Amsterdam, Netherlands

Department of Hematology, Amsterdam University Medical Centers, Cancer Center Amsterdam, Location VUmc, Amsterdam, Netherlands

In the rapidly developing field of adoptive cell immunotherapy, there is urgent need for discoveries that would improve outcomes, extend the applicability, and reduce the costs. Induced pluripotent stem cells (iPSC) can be a source of broadly applicable cellular immunotherapeutics, which have been manufactured, validated, and banked in advance, and can be applied across HLA barriers. Here, we discuss the recent advances and challenges in the generation of iPSC-derived cellular products for cancer therapy.

iPSCs can be differentiated to functional tumor-specific T and NK cells in vitro with demonstrable in vitro and in vivo anti-tumor activity. Genetic modifications employed at the iPSC level can deliver desirable immunotherapeutic attributes to the generated immune effectors. iPSC-NK cells are currently evaluated in a clinical setting and pre-clinical testing of iPSC-T cells shows promising results but their production seems more challenging.

The use of iPSCs for the generation of tumor-targeting T/NK cells constitutes a feasible strategy to overcome limitations in manufacturing, efficacy, and applicability of cellular therapeutics.

Keywords: Induced pluripotent stem cells, Adoptive cell immunotherapy, NK cells, T cells, Off-the-shelf

Adoptive cell immunotherapy for the treatment of hematological malignancies has greatly advanced in the last decades, leading to significant clinical outcomes. Both T cells and NK cells have been proven robust therapeutic agents and several approaches to obtain anti-tumor therapeutic lymphocytes have been proposed with variable clinical impact. These include the isolation and expansion of patient-derived tumor antigen-specific T cells (T cell clones, tumor infiltrating lymphocytes), innate T cells (NK-T or -T cells), or NK cells [1, 2]. The introduction of genetic engineering with T cell receptors (TCR) and most importantly chimeric antigen receptors (CAR) boosted the applicability and efficacy of adoptive cell immunotherapy [3]. CARs are artificial receptors that redirect antigen recognition from T cells and mediate T cell activation, through the fusion of an extracellular antigen-binding moiety, such as a single-chain-variable region (scFv), with an intracellular signaling domain [4]. CAR-modified T cells (CAR-T) targeting CD19 have induced impressive responses in chemotherapy-resistant B cell leukemias and lymphomas [5, 6], while B cell maturation antigen (BCMA)targeting CAR-T cells show promising clinical results against multiple myeloma [7]. These remarkable results led further to the development of CAR-engineered NK cells (CAR-NK), redirecting the intrinsic capacity of NK cells for tumor recognition and elimination and the initiation of CAR-NK clinical trials [8, 9].

Although successful, current approaches of adoptive cell therapy have significant limitations that impede their further progress and broader use. Immunotherapy using primary T cells is mainly performed in an autologous setting limiting a facile and general use. The production of genetically engineered therapeutic cells is a time-consuming process and the required processing time can be detrimental for the patients health. Also, in many cases, the autologous T cell isolation and expansion could be problematic and their functionality and quality doubtful (e.g., patients receiving highly immunosuppressive therapy). In addition, the existing ex vivo T cell expansion protocols push T cells to a terminal differentiated effector state, resulting in exhausted, less effective cellular products. The use of allogeneic cells from volunteer donors has the potential to broaden the applicability of the T cell products to HLA-matched recipients [10]. The advances of gene editing technology allow for further broadening of the applicability of CAR-T cell therapy by the use of allogeneic volunteer donor T cells that have been modified to lack TCR expression and thus, avoid graft-versus-host disease (GvHD) [10]. However, the final purity of the TCR-less CAR-T cell product is not always acceptable. Carriage of even <1% (<45104 cells/kg) of residual TCR-expressing T cells was still enough to initiate GvHD in the first clinical application of TCR-less CAR-T cells [10]. In addition, the potential off-target genetic alterations require further safety testing of every different batch produced that brings higher production costs. Extra gene editing to also eliminate HLA class I expression and reduce the risk of rejection could further reduce production efficiency (60% of double targeting efficiency) [10, 11] and require extra ex vivo expansion in order to reach the desirable yield of fully edited cells. However, it is known that longer ex vivo culture can be at the cost of having a more exhausted final product [12]. Notably, NK cells exert their function regardless of the recipients HLA haplotype and thus they can be isolated from an unrelated donor or cord blood. Clinical trials using adoptively transferred allogeneic NK cells showed limited toxic side effects such as GvHD [1317]. However, additionally to the above-mentioned limitations, their inadequate proliferative capacity impedes their in vivo persistence without cytokine support [18] and makes multiplex gene editing challenging. Immortalized NK cell lines, such as NK92, have been used as an alternative but they have to be irradiated before being infused to the patient [9, 19]. This limits their in vivo survival and thus, multiple doses of high numbers of NK92 cells are required [9]. Clinical application of irradiated NK92 cells transduced with a CD33-CAR was safe but without impressive anti-tumor results [9].

The development of broadly applicable cellular therapeutics, which have been manufactured, functionally validated and banked in advance, and can be applied across HLA barriers would improve the consistency and availability and reduce the cost of adoptive cell therapy. The generation of human lymphocytes from iPSCs has attracted lately the interest of the scientific community since it offers tantalizing prospects for cell-based therapies serving as an endless supply of custom-made, off-the-shelf therapeutic lymphocytes. Here, we review the latest advances in generating therapeutic anti-tumor T and NK lymphocytes from human induced pluripotent stem cells (iPSCs) and future challenges towards the final development of universally applicable immunotherapeutic products.

The iPSC technology offers new perspectives for the production of immunotherapeutic cellular products due to two major characteristics. First, similar to embryonic stem cells, iPSCs can be cultured unlimitedly in vitro and be successfully differentiated towards the lymphoid lineage [20]. Having access to constant and continuous production of T and/or NK lymphocytes offers solution to cell number and doses limitations due to restricted availability or expansion of primary cells. Second, iPSCs can be easily amenable to genetic transformations in vitro and thus, can generate immune effectors, which may eventually be genetically modified to augment their applicability, potency, and persistence. While the potential for multiplex gene editing is limited in primary cells, iPSC can theoretically bear unlimited genetic changes. Finally, in contrast to primary cells [10], genetic engineering of iPSCs results in fully modified clonal lines, which could be extensively evaluated resulting in a stable and safe source. The generation of safe master iPSC lines, bearing genetic modifications that confer the desired characteristics to the final product, would facilitate the development of off-the-shelf cellular therapeutics for more patients and types of malignancy.

Up to date almost all somatic cell types have been successfully reprogrammed into iPSCs through introduction of defined transcription factors (Oct4, Sox2, Klf4, c-Myc or Oct4, Sox2, Nanog, Lin28) [21, 22]. However, the selection of the initial iPSC source seems to be important when aiming in the efficient generation of therapeutically relevant T or NK lymphocytes (Fig.). Although, iPSC, from any somatic cell, can be successfully differentiated towards the lymphoid lineage, it has been shown that starting from blood cell-derived iPSCs, such as CD34+ cells from cord blood, monocytes or peripheral blood lymphocytes, results in a more efficient generation of CD4+CD8+ double-positive (DP) thymocytes in vitro [23] suggesting a level of epigenetic memory [24]. Moreover, iPSCs from peripheral blood T lymphocytes (T-iPSC) have the unique characteristic of bearing the rearranged TCR loci of the parental cells, which remain unchanged during in vitro differentiation [21, 23]. Therefore, T cells with defined TCR-specificity (e.g., T cell clones, invariant T cells, NK-T cells) can be selected for reprogramming to T-iPSC for therapeutic T cell production.

a Peripheral blood cells serve as a primary source to generate iPSC lines by non-integrating delivery of reprogramming transcription factors. b Generation of iPSC-derived off-the-shelf tumor-specific T cells. iPSCs are genetically modified to bear desirable immunotherapeutic properties. The expression of TCR and HLA is knocked out or silenced to prevent alloreactivity and graft rejection respectively. HLA-E/G molecules can be overexpressed to avoid NK cellmediated transplant rejection, whereas antigen-specific TCR/CARs can direct anti-tumor activity. Further, introduction of the expression of immune receptors, cytokines, chemokines, or other immune regulatory factors may enhance anti-tumor function. Genome-edited master iPSC lines are differentiated under GMP-grade conditions to fully functional histocompatible tumor-targeting T cells accessible to all patients regardless of their HLA haplotype

The hallmark of adoptive T cell immunotherapy is the use of the ability of T cells to specifically recognize tumor antigens. Naturally, this ability is endowed through the expression of specific TCRs encoded by the uniquely rearranged genomic loci of the TCR and chains. Generation of T cells from embryonic stem cells or iPSC, which bear non-rearranged germline TCR loci, results in random rearrangements during differentiation and a population of cells with various unknown specificities [25]. Nishimura et al. demonstrated that differentiation of T-iPSCderived from an antigen-specific T cell clone gives rise to T cells with the same TCR rearrangement and reactivity [21], thus making T-iPSC from tumor antigen-specific T cells a way to deliver defined specificity to iPSC-derived T cells. Until now, T cell clones specific for several tumor antigens, such as MART-1 (melanoma), LMP2 (EBV antigen), WT-1 (leukemia), and GPC3 (hepatocellular, ovarian, and lung carcinoma), have been reprogrammed to T-iPSC [23, 26, 27]. Importantly, tumor-specific T-iPSC could give rise to cytotoxic CD8 single-positive (SP) T cells, which could recognize the target antigen on cell lines and display specific cytotoxicity [23, 26, 27]. Although most of the generated cells bear the original tumor-specific TCR, it has been observed that TCR can be additionally rearranged, resulting in TCR destabilization and loss of the antigen specificity [26]. Inactivation of recombination activating gene 2 (RAG2) in the T-iPSC, a key protein complex in the rearrangement of TCR, can inhibit the process of further TCR rearrangement at the DP stage and result in preservation of antigen specificity in CD8 T cells [26].

Although taking advantage of a naturally occurring TCR in order to convey antigen specificity to iPSC-derived T cells, one can imagine that the flexibility of this method is limited, as the existence of pre-made, HLA-matched antigen-specific T cell clones is a requirement. The advances in engineering of immune cells open up new perspectives for the generation of custom-made synthetic T cells from iPSC. Antigen specificity can be assigned to iPSCs and their T cell derivatives by means of a transgenic TCR [26]. Themeli et al. demonstrated the feasibility of generating functional CAR-T cells by engineering T-iPSC with a CAR [28]. The use of CARs endows T-iPSCderived T cells with HLA-independent, customizable antigen recognition as scFv domains of different specificity can be used giving the potential for a wider applicability range. Importantly, second- and third-generation CARs provide additionally costimulatory signals and enhance T cell activation, expansion, and in vivo persistence [4]. In principal, similarly to conventional CAR-T cells, T-iPSC can be also further genetically armed with cytokines, receptors, and other regulatory molecules in order to provide their derivatives with optimal immunotherapeutic properties such as enhanced proliferation and reduced exhaustion [2932]. Especially the rise of gene editing technologies, such as CRISPR/Cas9 and TALEN technologies, offers new perspectives for the multiplex modification of T-iPSC.

The potential of iPSCs to become a valuable source of readily available anti-tumor T cells depends on the development of a defined and efficient production process that could yield the cell numbers required for clinical application. In 2009, Timmermans et al. first reported the derivation of mature CD3+TCR+ T cells from human embryonic stem cells [25]. Since then, several groups have demonstrated successful in vitro generation of T lymphocytes from iPSC, although using slightly different methods [21, 23, 28]. However, all described protocols follow the same differentiation path, recapitulating the process of human T cell development. First, iPSCs are induced to form mesoderm from which definitive hemogenic endothelium arises as a next step. Hemogenic endothelium is then transitioning to form a pool of hematopoietic stem and progenitor cells (HSPC), a subpopulation of which has the potential to commit to the T cell lineage. The final important steps involve the emergence of CD8+/CD4+ DP cells and eventually of mature CD8 or CD4 SP T lymphocytes. Since this is a multi-step differentiation process where every developmental transition happens with different efficiency, one could imagine that the production of mature SP T cells for clinical applications is a challenge. In recent studies, investigators were able to generate enough numbers of anti-tumor T cells in order to test their functionality in vivo in xenograft murine models [26, 27, 28]. However, the development of an efficient method for the production of clinically relevant cell numbers is still not reported.

Another major challenge is that, although successful in generating T cells from iPSC or T-iPSC, most of the reported differentiation methods include the presence of uncharacterized serum and feeder cells of murine origin, which are not compatible with clinical level production. Induction of the hematopoietic program has been achieved through co-culture with OP9, a murine bone marrow stromal cell line, or C3H10T1/2, a mouse embryonic fibroblast line [21, 23, 33]. Further T lymphoid commitment requires the use of the same cell lines of murine origin transduced to overexpress the Notch ligands DLL1 or DLL4 [20, 21, 23, 33]. One could replace the murine feeder cells with cells of human origin, but until now attempts to create human-origin feeder cells for T cell development had disappointing results, as human fibroblasts or keratinocytes have failed to efficiently support the differentiation of human CD34+ to pro T cells or SP mature cells [34, 35]. Therefore, the development of a feeder-free and serum-free method is required. Kennedy et al. managed to replace feeder use, for the differentiation of iPSC towards CD34+CD43 HSPC, with a serum-free and stroma cell-free protocol based on embryoid body formation and the use of rationally selected cytokine combinations in a stepwise manner [20]. Creating an in vitro thymic niche by using plate-bound recombinant molecules of DLL4/DLL1 and VCAM-1, fused to the Fc portion of human IgG, has been used to generate T cell lineage cells from or cord blood-derived CD34+ cells [36], but the successful feeder-free differentiation of iPSC-derived HSPC has not been described.

Beyond antigen specificity, the functional potential of T cells depends also on their developmental maturity and their lineage subtype (, , -, CD4 or CD8, Treg, etc). This underscores once more the importance of developing differentiation protocols, which are based on the knowledge of human T lymphoid development, for the generation of the T cell subtype with the desired functionality. The first studies generating cytotoxic T cells from T-iPSC revealed that their phenotype and functionality was not precisely similar to that of mature CD8 T cells. The T-iPSCderived rejuvenated T cells although they were CD3+TCR+ showed T cellspecific gene expression profile and elicited specific cytotoxic responses against cells expressing the target antigen; they lacked expression of important surface molecules (such as CD2, CD5, CD28) and expressed high levels of innate T cell-related markers (such as CD56) [21, 23]. Also, Themeli et al. further reported that CAR-modified T-iPSC differentiate into CD3+TCR+CD8+ CAR-T cells whose gene expression profile was similar to that of -T cells [28]. Importantly, their in vivo anti-tumor functionality was analogous to that of peripheral blood-derived -T cells from the same donor bearing the same CAR [28]. Therefore, although the generated T-iPSCderived T cells express the endogenous TCR, they have phenotypic and functional characteristics of an innate-like lymphocyte. Similar lineage skewing has been observed in transgenic TCR mice [3739] and in vitro differentiation of TCR-engineered human CD34+ hematopoietic progenitors [40], wherein the emerging TCR+ T cells displayed innate T cell features, such as expression of CD8 and low levels of CD5 [37]. CD8 is expressed as a CD8 heterodimer on mature cytotoxic T cells while CD8 homodimers are present only on innate lymphocyte subtypes such as NK cells, -T cells, NK-T cells, or intestinal epithelial lymphocytes (IEL) [41, 42]. Heterodimeric CD8 has been shown to be a better coreceptor for TCR/pMHC binding than homodimeric CD8 [43] and the presence of CD8 is indicative of maturity. Indeed, T-iPSCderived T cells expressing CD8 exhibited improved antigen-specific cytotoxicity in vitro and in vivo compared with CD8 cells which show innate-like non-antigen-specific reactivity [27].

It has been suggested that premature expression of the transgenic TCR may prevent -selection of the cells, skew development towards the lineage, and result in the emergence of TCR-expressing T cells with properties [37, 38]. Interestingly, the pre-rearranged endogenous TCR of T-iPSC is already expressed on day 1520 of differentiation on OP9-DL1 [28], which is remarkably earlier than the appearance of TCR/ in differentiation of cord blood (CB)-CD34+ cells and some other reports on human ES/iPSC T cell differentiation [20, 25]. In addition, similarly to what is reported for the TCR transgenic mice, the generation of CD8-expressing DP cells from T-iPSC is very inefficient [21, 27, 33]. Interestingly, previous studies in transgenic murine models have demonstrated that lineage determination during T lymphoid differentiation is dependent on the synergy between TCR and Notch signaling and differences in Notch signal strength are also an important factor influencing versus development [44]. Murine T-iPSC differentiated in a 3D thymic culture generated antigen-specific anti-tumor T cells, which were phenotypically and functionally more similar to nave CD8 T cells in contrast to CD8 cells emerging from the OP9-DL1 co-culture [45]. Therefore, a thymic environment provides the correct combination of Notch and other signals that promote the maturation of thymocytes. Further research should focus on the development of in vitro differentiation systems that better mimic the interactions and the balanced TCR and Notch signaling that takes place within the thymus.

Phenotypic and functional evaluation of the T-iPSCderived CD8 T cells [27] or iPSC-derived CD8 T cells bearing a transgenic TCR [26] showed that they are similar to their peripheral blood counterparts. However, there are still gene expression discrepancies such as the lack of chemokine receptors (CCR7, CXCR3) and a weaker but still existing NK-like cytotoxicity [27]. In addition, it is still not clear whether their anti-tumor function is equivalent to that of conventional CD8 T cells. The in vivo anti-tumor functionality of regenerated CD8 T cells has been up to date tested in xenograft models where the tumor cells are inoculated intraperitoneally or subcutaneously and thus, not in a naturally occurring location limiting our insight on their migratory capacities [26, 27, 28, 33]. Finally, although treatment with iPSC-derived CD8 T cells significantly delayed tumor growth, it required multiple injections of high numbers of cells [26, 27, 33]. The use of xenograft models where tumor cells are inoculated at their natural sites and T cell-dose escalation could be more informative on the functionality of the iPSC-derived T cells.

NK cells constitute a robust part of the innate immune system implicated in recognition and lysis of malignant and virally infected cells. Their HLA-independent cytotoxic capacity makes them favorable candidates for off-the-shelf cellular therapeutic product compared with T cells. However, as already mentioned, their limited proliferative and genetic manipulation potential render their clinical application challenging. The perspective of manufacturing therapeutic NK cells from iPSC provides solutions to many of the bottlenecks of adoptive NK cell therapy.

Interestingly, the generation of NK cells from iPSC has proven easier and more straight-forward than the production of antigen-specific cytotoxic T cells. Although the first steps of differentiation towards HSPCs are similar to that of T differentiation, the commitment to NK lymphoid lineage is less complicated and does not require the presence of Notch signaling. Several studies have demonstrated the robust production of homogeneous, mature NK cells from human iPSC, which express all significant NK-defining markers such as CD56, FcRIIIa receptor (CD16), CD94, killer immunoglobulin-like receptors (KIRs), natural cytotoxicity receptors (NKp30, NKp44, and NKp46), activating receptors (NKG2D and DNAM-1), and death-inducing ligands (FasL, TRAIL) [46, 47, 48]. Most importantly, Knorr et al. reported a clinical-grade, serum-free, and feeder-free differentiation protocol to obtain functional NK cells from iPSC, which involves embryoid body formation in defined conditions and the use of membrane-bound interleukin 21-expressing artificial antigen-presenting cells [49]. According to the authors of the study, enough cytotoxic NK cells to treat a single patient could be produced from fewer than 250,000 input hiPSCs, thus facilitating the potential for clinical application in cancer therapy.

Apart from a mature phenotype, iPSC-derived NK cells display efficient cytotoxic capacity through direct receptor-mediated lysis, cytokine and chemokine secretion, and antibody-dependent cell-mediated cytotoxicity (ADCC) [46, 47]. When evaluating the anti-tumor activity in an ovarian cancer xenograft model, intraperitoneally injected iPSC-derived NK cells showed similar delay of tumor progression and overall survival as peripheral blood NK cells expanded on artificial antigen-presenting cells (aAPC) [47]. However, although statistically significant, this anti-tumor effect was not impressive and required multiple doses of NK cells. Interestingly, engineering iPSCs with a CAR bearing NK-specific costimulatory domains derived from NKG2D and 2B4 proteins optimized the targeted anti-tumor activity of the generated CARiPSC-NK cells and improved their in vivo expansion and cytotoxic capacity [48]. Importantly, a single dose of CARiPSC-NK cells resulted in less toxicity but similar anti-tumor effect as third-generation CAR-T cells against ovarian cancer in vivo, although comparison of tumor burden was limited to 3weeks post infusion in this study [48]. Further genetic engineering has been proposed in order to improve the function and therapeutic potential of iPSC-NK cells. For example, the expression of a non-cleavable CD16 would enhance ADCC potential, the addition of an IL15R-IL-15 fusion can provide self-stimulation, and expression of CXCR3 can improve homing of iPSC-NK cells [50].

Although generation of T and NK cells from iPSC overcomes many of the limitations of current manufacturing practices, their use would only really facilitate the applicability of adoptive cell therapy if they are available as a true universally applicable off-the-shelf product, which could be infused to any patient.

The major barrier limiting the applicability of allogeneic iPSC-derived products is the HLA-disparity between the effector T cells and the host, which may lead to graft rejection or graft-versus-host (GvH) reaction. Using matched previously banked iPSCs from HLA-homozygous donors as a starting material has been previously proposed [51]. It has been calculated that an iPSC bank with 50 HLA-homozygous iPSC lines could cover approximately 73% of the Japanese population [51] while 93% of the UK population would find a match within 150 HLA-homozygous lines [52]. However, the establishment of universal iPSC lines is considered to provide a true solution as they would provide widely applicable cellular products without the need for HLA-matching. Hypoimmunogenic, histocompatible pluripotent stem cell lines can be generated by elimination of HLA class I and II expression by disruption of 2m and CIITA gene respectively [53, 54, 55]. Allogeneic cells, which lack self class I HLA molecules, can however be rejected by host NK cells. Introduction of HLA-E, HLA-G, or of patient-specific HLA-C has been shown to reduce NK-mediated rejection of iPSC-derived cells [53, 54, 55]. Introduction of additional immunomodulatory molecules such as PD-L1 and CD47 can further reduce the recipients immune responses [55].

When using allogeneic T cells, the possibility of graft-versus-host reactions is a major concern. In order to avoid alloreactivity of iPSC-derived T cells, T-iPSC bearing an endogenous TCR of known specificity (virus- or cancer-specific) could be used. Alternatively, the surface expression of the TCR could be disrupted by the means of genome editing as previously described for conventional CAR-T cells [10, 56].

Although, many of the above genetic modifications have been already reported and evaluated in the iPSC level, there is up to date no study showing the generation of functional and mature lymphocytes from universal iPSCs.

The advancement of adoptive cell immunotherapy and the impressive clinical outcomes obtained targeting hematologic malignancies with CAR-T cells dictate for further developments towards a broader use of cellular therapeutics for more patients and more types of malignancy. The advent of iPSC technology provides new perspectives for the manufacturing of customized, tumor-targeting T/NK cells, with improved immunotherapeutic properties and the potential of universal off-the-shelf use (Fig. ). Rapid progress in the field of lymphoid differentiation of iPSC has brought the clinical application of iPSC-derived adoptive immunotherapy from theory to reality. Indeed, the first clinical trial testing an off-the-shelf, iPSC-derived NK cell product against advanced solid tumors started recruiting in 2019 (ClinicalTrials.gov Identifier: {"type":"clinical-trial","attrs":{"text":"NCT03841110","term_id":"NCT03841110"}}NCT03841110). In addition, the production of iPSC-derived T cells and TCR/CAR-engineered T cells is already in pre-clinical development. Fate Therapeutics is developing TCR-less T-iPSCderived CD19-CAR-T cells where the CD19CAR is expressed from the TCR chain constant region (TRAC) locus, while Adaptimmune aims to develop off-the-shelf anti-tumor T cells from TCR-engineered iPSC.

However, there are still several challenges to be pre-clinically addressed before the first clinical application of iPSC-derived T cells. As mentioned above, the phenotypic and functional maturity of the generated T cell effectors has to be ensured as well as an anti-tumor potential comparable with natural T cells. Further, manufacturing protocols should be established which would allow for the efficient, GMP-grade, and clinical scale production of iPSC-derived T cell products. Finally, as with all iPSC-derived cellular products, the potential risk of malignant transformation due to contamination with undifferentiated iPSC has to be minimized, for example with the use of suicide genes such as the iC9/CID system [33]. Further future advances in iPSC and genome editing technologies in combination with in-depth knowledge of the fundamental mechanisms of T/NK cell function and the regulation of lymphoid development will provide the tools for the generation of iPSC-derived T/NK cell products with improved therapeutic anti-tumor function, better homing, persistence, and applicable across histocompatibility barriers.

Alexandros Nianias declares that he has no conflict of interest.

Maria Themeli reports serving as consultant for Covagen AG. In addition, Dr. Themeli has a patent WO2014165707A2 with royalties paid to Fate Therapeutics.

This article does not contain any studies with human or animal subjects performed by any of the authors.

This article is part of the Topical Collection on CART and Immunotherapy

Publishers Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Alexandros Nianias, Email: ln.cmumadretsma@sainain.a.

Maria Themeli, Phone: +31 (0) 204447413, Email: ln.cmumadretsma@ilemeht.m.

View original post here:
Induced Pluripotent Stem Cell (iPSC)Derived Lymphocytes for Adoptive ...