Category Archives: Stem Cell Treatment


ASCO Expert Discusses the Use of Autologous Transplantation in Relapsed DLBCL – Cancer Network

In an interview with CancerNetwork, Nirav Niranjan Shah, MD, of the Medical College of Wisconsin, discussed the use of autologous stem cell transplant in patients with relapsed chemosensitive diffuse large B-cell lymphoma (DLBCL) during the era of CAR T-cell therapy.

In a data analysis using the Center of International Bone Marrow Transplant Registry (CI-BMTR), Shah and fellow investigators evaluated whether the use of autologous transplantation in this patient population should remain the standard of care.

The results of the analysis, presented at the 2020 American Society of Clinical Oncology (ASCO) Virtual Scientific Meeting, strongly supported that autologous transplantation should remain the current standard of care.

Transcription:

Yes, I think the most important finding that I take away from this is that until we have randomized controlled trial data that shows that CAR T-cell is superior to autologous stem cell transplant in this specific patient population; that autologous transplant is the standard of care. Obviously, if you have chemo-refractory disease, well, that's a different patient population and those patients should be receiving CAR T-cell therapy. But I think we more need more data before we outright say that autologous transplant is no longer the most appropriate therapy for that particular population.

You know, the other major finding we found is that the overall survival did favor patients who had late chemotherapy failure. So that was a finding that we saw in our multivariate analysis, but besides that point, there was no difference in transplant related mortality rates of relapse or progression. And there's no difference in progression-free survival, which were the other endpoints of our analysis.

Original post:
ASCO Expert Discusses the Use of Autologous Transplantation in Relapsed DLBCL - Cancer Network

Adipose Tissue-derived Stem Cell Therapy Market 2020 | by Manufacturers | by Countries | by Types and by Applications | by Forecasts to 2026 – Farmers…

The Adipose Tissue-derived Stem Cell Therapy Market report we provide to our readers contains comprehensive data on a specific product/service, available in this industry. We want to perform in-depth analysis, to obtain a comprehensive understanding of the Adipose Tissue-derived Stem Cell Therapy Market. It starts off by going to the basics of the product/service, which is to take a look at the industry definition. The Adipose Tissue-derived Stem Cell Therapy Market report identifies and analyzes the factors which contribute and hamper the growth of this line of business. At the same time, we identify the current value of the Adipose Tissue-derived Stem Cell Therapy Market, with the estimated financial worth, at the end of the forecast period, 2020-2026.

One metric we use to understand the potential growth of the Adipose Tissue-derived Stem Cell Therapy Market is to calculate the CAGR. It helps provide accurate data, improving the quality of the data collected for this report. We make sure to analyze all the information available in this document, to ensure it meets our standards. In this report, the reader will learn which elements are responsible for creating demand for the product/service under observation. At the same time, the reader will also get to know about product/service types that boost the popularity of this industry.

The key players covered in this study > AlloCure, Antria, Celgene Corporation, Cellleris, Corestem, Cytori Therapeutics, Intrexon, Mesoblast, Pluristem Therapeutics, Tissue Genesis, BioRestorative Therapies, Celltex Therapeutics Corporation, iXCells Biotechnologies, Pluristem Therapeutics, Cyagen, Lonza.

The final report will add the analysis of the Impact of Covid-19 in this report Adipose Tissue-derived Stem Cell Therapy industry.

Get a Sample Copy @ https://www.reportsandmarkets.com/sample-request/covid-19-impact-on-global-adipose-tissue-derived-stem-cell-therapy-market-size-status-and-forecast-2020-2026-one

Market Segmentation

For the purpose of making the information available on Adipose Tissue-derived Stem Cell Therapy Market comprehensive, we segmented the industry. The reason is that it helps our readers learn in-depth about this line of business. The segmentation of the Adipose Tissue-derived Stem Cell Therapy Market is as follows distribution channel, product type, region, and application. When it comes to application, it deals with end-users, who are responsible for generating demand for the product/service. Product type refers to the different variants available in the Adipose Tissue-derived Stem Cell Therapy Market. We use distribution channel, to understand the various sources companies use to supply the product/service to the consumers.

Regional Overview

In the regional overview portion, the Adipose Tissue-derived Stem Cell Therapy Market report has data from countries all over the world. Each region is responsible for contributing to the growth of this industry. From the available data, we will identify which area has the largest share of the market. At the same time, we will compare this data to other regions, to understand the demand in other countries. North and South America, Asia Pacific, Middle East and Africa, and Europe are the areas of interest in this Adipose Tissue-derived Stem Cell Therapy Market report.

Table Of Content

1 Report Overview

2 Global Growth Trends

3 Market Share by Key Players

4 Breakdown Data by Type and Application

5 North America

6 Europe

7 China

8 Japan

9 Southeast Asia

10 India

11 Central & South America

12 International Players Profiles

13 Market Forecast 2020-2026

14 Analysts Viewpoints/Conclusions

15 Appendix

know more about this report @ https://www.reportsandmarkets.com/enquiry/covid-19-impact-on-global-adipose-tissue-derived-stem-cell-therapy-market-size-status-and-forecast-2020-2026-one

Latest Industry News

We will cover government policies, which favor or go against the Adipose Tissue-derived Stem Cell Therapy Market, as we believe this can change the level of growth. At the same time, technological advancements which have the power to influence the growth will appear in the latest industry news.

Any special requirements about this report, please let us know and we can provide custom report.

About Us:

Market research is the new buzzword in the market, which helps in understanding the market potential of any product in the market. Reports And Markets is not just another company in this domain but is a part of a veteran group called Algoro Research Consultants Pvt. Ltd. It offers premium progressive statistical surveying, market research reports, analysis & forecast data for a wide range of sectors both for the government and private agencies all across the world.

For more detailed information please contact us at:

Sanjay Jain

Manager Partner Relations & International Marketing

http://www.reportsandmarkets.com

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

Originally posted here:
Adipose Tissue-derived Stem Cell Therapy Market 2020 | by Manufacturers | by Countries | by Types and by Applications | by Forecasts to 2026 - Farmers...

Trending 2020 Lysosomal Storage Disease Treatment Market Segmentation, Analysis by Recent Trends, Development & Growth by Regions – Cole of Duty

Lysosomal Storage Disease TreatmentMarket 2020: Inclusive Insight

Los Angeles, United States, June 2020:The report titled Global Lysosomal Storage Disease Treatment Market is one of the most comprehensive and important additions to Alexareports archive of market research studies. It offers detailed research and analysis of key aspects of the global Lysosomal Storage Disease Treatment market. The market analysts authoring this report have provided in-depth information on leading growth drivers, restraints, challenges, trends, and opportunities to offer a complete analysis of the global Lysosomal Storage Disease Treatment market. Market participants can use the analysis on market dynamics to plan effective growth strategies and prepare for future challenges beforehand. Each trend of the global Lysosomal Storage Disease Treatment market is carefully analyzed and researched about by the market analysts.

Lysosomal Storage Disease Treatment Market competition by top manufacturers/ Key player Profiled: Takeda, Pfizer, Sanofi, BioMarin, Merck, Actelion Pharmaceuticals, Eli Lilly,

Get PDF Sample Copy of the Report to understand the structure of the complete report:(Including Full TOC, List of Tables & Figures, Chart) : https://www.alexareports.com/report-sample/856284

Global Lysosomal Storage Disease Treatment Market is estimated to reach xxx million USD in 2020 and projected to grow at the CAGR of xx% during 2020-2026. According to the latest report added to the online repository of Alexareports the Lysosomal Storage Disease Treatment market has witnessed an unprecedented growth till 2020. The extrapolated future growth is expected to continue at higher rates by 2026.

Lysosomal Storage Disease Treatment Market Segment by Type covers: Enzyme Replacement Therapy, Stem Cell Transplantation, Substrate Reduction Therapy

Lysosomal Storage Disease Treatment Market Segment by Application covers:Hospitals, Clinics, Stem Transplant Centers, Research Organizations

After reading the Lysosomal Storage Disease Treatment market report, readers get insight into:

*Major drivers and restraining factors, opportunities and challenges, and the competitive landscape*New, promising avenues in key regions*New revenue streams for all players in emerging markets*Focus and changing role of various regulatory agencies in bolstering new opportunities in various regions*Demand and uptake patterns in key industries of the Lysosomal Storage Disease Treatment market*New research and development projects in new technologies in key regional markets*Changing revenue share and size of key product segments during the forecast period*Technologies and business models with disruptive potential

Based on region, the globalLysosomal Storage Disease Treatment market has been segmented into Americas (North America ((the U.S. and Canada),) and Latin Americas), Europe (Western Europe (Germany, France, Italy, Spain, UK and Rest of Europe) and Eastern Europe), Asia Pacific (Japan, India, China, Australia & South Korea, and Rest of Asia Pacific), and Middle East & Africa (Saudi Arabia, UAE, Kuwait, Qatar, South Africa, and Rest of Middle East & Africa).

Key questions answered in the report:

What will the market growth rate of Lysosomal Storage Disease Treatment market?What are the key factors driving the global Lysosomal Storage Disease Treatment market size?Who are the key manufacturers in Lysosomal Storage Disease Treatment market space?What are the market opportunities, market risk and market overview of the Lysosomal Storage Disease Treatment market?What are sales, revenue, and price analysis of top manufacturers of Lysosomal Storage Disease Treatment market?Who are the distributors, traders, and dealers of Lysosomal Storage Disease Treatment market?What are the Lysosomal Storage Disease Treatment market opportunities and threats faced by the vendors in the global Lysosomal Storage Disease Treatment industries?What are sales, revenue, and price analysis by types and applications of Lysosomal Storage Disease Treatment market?What are sales, revenue, and price analysis by regions of Lysosomal Storage Disease Treatment industries?

GetExclusive discount on this report now at https://www.alexareports.com/check-discount/856284

Table of ContentsSection 1 Lysosomal Storage Disease Treatment Product DefinitionSection 2 Global Lysosomal Storage Disease Treatment Market Manufacturer Share and Market Overview2.1 Global Manufacturer Lysosomal Storage Disease Treatment Shipments2.2 Global Manufacturer Lysosomal Storage Disease Treatment Business Revenue2.3 Global Lysosomal Storage Disease Treatment Market Overview2.4 COVID-19 Impact on Lysosomal Storage Disease Treatment IndustrySection 3 Manufacturer Lysosomal Storage Disease Treatment Business Introduction3.1 Takeda Lysosomal Storage Disease Treatment Business Introduction3.1.1 Takeda Lysosomal Storage Disease Treatment Shipments, Price, Revenue and Gross profit 2014-20193.1.2 Takeda Lysosomal Storage Disease Treatment Business Distribution by Region3.1.3 Takeda Interview Record3.1.4 Takeda Lysosomal Storage Disease Treatment Business Profile3.1.5 Takeda Lysosomal Storage Disease Treatment Product Specification3.2 Pfizer Lysosomal Storage Disease Treatment Business Introduction3.2.1 Pfizer Lysosomal Storage Disease Treatment Shipments, Price, Revenue and Gross profit 2014-20193.2.2 Pfizer Lysosomal Storage Disease Treatment Business Distribution by Region3.2.3 Interview Record3.2.4 Pfizer Lysosomal Storage Disease Treatment Business Overview3.2.5 Pfizer Lysosomal Storage Disease Treatment Product Specification3.3 Sanofi Lysosomal Storage Disease Treatment Business Introduction3.3.1 Sanofi Lysosomal Storage Disease Treatment Shipments, Price, Revenue and Gross profit 2014-20193.3.2 Sanofi Lysosomal Storage Disease Treatment Business Distribution by Region3.3.3 Interview Record3.3.4 Sanofi Lysosomal Storage Disease Treatment Business Overview3.3.5 Sanofi Lysosomal Storage Disease Treatment Product Specification3.4 BioMarin Lysosomal Storage Disease Treatment Business Introduction3.5 Merck Lysosomal Storage Disease Treatment Business Introduction3.6 Actelion Pharmaceuticals Lysosomal Storage Disease Treatment Business IntroductionSection 4 Global Lysosomal Storage Disease Treatment Market Segmentation (Region Level)4.1 North America Country4.1.1 United States Lysosomal Storage Disease Treatment Market Size and Price Analysis 2014-20194.1.2 Canada Lysosomal Storage Disease Treatment Market Size and Price Analysis 2014-20194.2 South America Country4.2.1 South America Lysosomal Storage Disease Treatment Market Size and Price Analysis 2014-20194.3 Asia Country4.3.1 China Lysosomal Storage Disease Treatment Market Size and Price Analysis 2014-20194.3.2 Japan Lysosomal Storage Disease Treatment Market Size and Price Analysis 2014-20194.3.3 India Lysosomal Storage Disease Treatment Market Size and Price Analysis 2014-20194.3.4 Korea Lysosomal Storage Disease Treatment Market Size and Price Analysis 2014-20194.4 Europe Country4.4.1 Germany Lysosomal Storage Disease Treatment Market Size and Price Analysis 2014-20194.4.2 UK Lysosomal Storage Disease Treatment Market Size and Price Analysis 2014-20194.4.3 France Lysosomal Storage Disease Treatment Market Size and Price Analysis 2014-20194.4.4 Italy Lysosomal Storage Disease Treatment Market Size and Price Analysis 2014-20194.4.5 Europe Lysosomal Storage Disease Treatment Market Size and Price Analysis 2014-20194.5 Other Country and Region4.5.1 Middle East Lysosomal Storage Disease Treatment Market Size and Price Analysis 2014-20194.5.2 Africa Lysosomal Storage Disease Treatment Market Size and Price Analysis 2014-20194.5.3 GCC Lysosomal Storage Disease Treatment Market Size and Price Analysis 2014-20194.6 Global Lysosomal Storage Disease Treatment Market Segmentation (Region Level) Analysis 2014-20194.7 Global Lysosomal Storage Disease Treatment Market Segmentation (Region Level) AnalysisSection 5 Global Lysosomal Storage Disease Treatment Market Segmentation (Product Type Level)5.1 Global Lysosomal Storage Disease Treatment Market Segmentation (Product Type Level) Market Size 2014-20195.2 Different Lysosomal Storage Disease Treatment Product Type Price 2014-20195.3 Global Lysosomal Storage Disease Treatment Market Segmentation (Product Type Level) AnalysisSection 6 Global Lysosomal Storage Disease Treatment Market Segmentation (Industry Level)6.1 Global Lysosomal Storage Disease Treatment Market Segmentation (Industry Level) Market Size 2014-20196.2 Different Industry Price 2014-20196.3 Global Lysosomal Storage Disease Treatment Market Segmentation (Industry Level) AnalysisSection 7 Global Lysosomal Storage Disease Treatment Market Segmentation (Channel Level)7.1 Global Lysosomal Storage Disease Treatment Market Segmentation (Channel Level) Sales Volume and Share 2014-20197.2 Global Lysosomal Storage Disease Treatment Market Segmentation (Channel Level) AnalysisSection 8 Lysosomal Storage Disease Treatment Market Forecast 2019-20248.1 Lysosomal Storage Disease Treatment Segmentation Market Forecast (Region Level)8.2 Lysosomal Storage Disease Treatment Segmentation Market Forecast (Product Type Level)8.3 Lysosomal Storage Disease Treatment Segmentation Market Forecast (Industry Level)8.4 Lysosomal Storage Disease Treatment Segmentation Market Forecast (Channel Level)Section 9 Lysosomal Storage Disease Treatment Segmentation Product Type9.1 Enzyme Replacement Therapy Product Introduction9.2 Stem Cell Transplantation Product Introduction9.3 Substrate Reduction Therapy Product IntroductionSection 10 Lysosomal Storage Disease Treatment Segmentation Industry10.1 Hospitals Clients10.2 Clinics Clients10.3 Stem Transplant Centers Clients10.4 Research Organizations ClientsSection 11 Lysosomal Storage Disease Treatment Cost of Production Analysis11.1 Raw Material Cost Analysis11.2 Technology Cost Analysis11.3 Labor Cost Analysis11.4 Cost OverviewSection 12 Conclusion

Do Inquiry About The Report Here: https://www.alexareports.com/send-an-enquiry/856284

About Us:Alexa Reports is a globally celebrated premium market research service provider, with a strong legacy of empowering business with years of experience. We help our clients by implementing a decision support system through progressive statistical surveying, in-depth market analysis, and reliable forecast data.

Contact Us:Alexa ReportsPh no: +1-408-844-4624Email: [emailprotected]Site: https://www.alexareports.com

Read the original:
Trending 2020 Lysosomal Storage Disease Treatment Market Segmentation, Analysis by Recent Trends, Development & Growth by Regions - Cole of Duty

Blood and Bone Marrow Cancer Treatment Market Overview Industry Demand, Development and Growth Forecast Report 2026 – Cole of Duty

QY Research has published a new report on the global Blood and Bone Marrow Cancer Treatment market is carried out by the analysts in this report, taking into consideration key factors like drivers, challenges, recent trends, opportunities, advancements, The analysts have clarified every part of the market exhaustive fastidious research and full focus to each point. This report gives additional information on statistical data to assist readers with understanding the entire market. This report offers a clear understanding of the present as well as future scenario of the global Blood and Bone Marrow Cancer Treatment industry. Research techniques like PESTLE and Porters Five Forces analysis have been deployed by the researchers. They have also provided accurate data on Blood and Bone Marrow Cancer Treatment production, capacity, price, cost, margin, and revenue to help the players gain a clear understanding into the overall existing and future market situation.

Key companies operating in the global Blood and Bone Marrow Cancer Treatment market include _ AstraZeneca, Plc., Celgene, Inc., Bristol Myers Squibb & Company, Eli Lilly & Company, Johnson & Johnson Company, F.Hoffman La-Roche Ltd., Merck & Co., Inc., Novartis AG, Pfizer, Inc., Varian Medical Systems, Inc. Blood and Bone Marrow Cancer Treatment Breakdown Data by Type

Get PDF Sample Copy of the Report to understand the structure of the complete report: (Including Full TOC, List of Tables & Figures, Chart) :

https://www.qyresearch.com/sample-form/form/1446786/global-blood-and-bone-marrow-cancer-treatment-market

Segmental Analysis

The report has classified the global Blood and Bone Marrow Cancer Treatment industry into segments including product type and application. Every segment is evaluated based on growth rate and share. Besides, the analysts have studied the potential regions that may prove rewarding for the Blood and Bone Marrow Cancer Treatment manufcaturers in the coming years. The regional analysis includes reliable predictions on value and volume, thereby helping market players to gain deep insights into the overall Blood and Bone Marrow Cancer Treatment industry.

Global Blood and Bone Marrow Cancer Treatment Market Segment By Type:

, Chemotherapy, Immunotherapy, Stem Cell Transplant, Radiotherapy Blood and Bone Marrow Cancer Treatment Breakdown Data by Application,

Global Blood and Bone Marrow Cancer Treatment Market Segment By Application:

Multiple Myeloma, Leukemia, Lymphoma, Others

Competitive Landscape

It is important for every market participant to be familiar with the competitive scenario in the global Blood and Bone Marrow Cancer Treatment industry. In order to fulfil the requirements, the industry analysts have evaluated the strategic activities of the competitors to help the key players strengthen their foothold in the market and increase their competitiveness.

Key companies operating in the global Blood and Bone Marrow Cancer Treatment market include _ AstraZeneca, Plc., Celgene, Inc., Bristol Myers Squibb & Company, Eli Lilly & Company, Johnson & Johnson Company, F.Hoffman La-Roche Ltd., Merck & Co., Inc., Novartis AG, Pfizer, Inc., Varian Medical Systems, Inc. Blood and Bone Marrow Cancer Treatment Breakdown Data by Type

Key questions answered in the report:

For Discount, COVID-19 Impact, Customization in the Report: https://www.qyresearch.com/customize-request/form/1446786/global-blood-and-bone-marrow-cancer-treatment-market

TOC

Table of Contents 1 Report Overview1.1 Study Scope1.2 Key Market Segments1.3 Players Covered: Ranking by Blood and Bone Marrow Cancer Treatment Revenue1.4 Market Analysis by Type1.4.1 Global Blood and Bone Marrow Cancer Treatment Market Size Growth Rate by Type: 2020 VS 20261.4.2 Chemotherapy1.4.3 Immunotherapy1.4.4 Stem Cell Transplant1.4.5 Radiotherapy1.5 Market by Application1.5.1 Global Blood and Bone Marrow Cancer Treatment Market Share by Application: 2020 VS 20261.5.2 Multiple Myeloma1.5.3 Leukemia1.5.4 Lymphoma1.5.5 Others1.6 Study Objectives1.7 Years Considered 2 Global Growth Trends by Regions2.1 Blood and Bone Marrow Cancer Treatment Market Perspective (2015-2026)2.2 Blood and Bone Marrow Cancer Treatment Growth Trends by Regions2.2.1 Blood and Bone Marrow Cancer Treatment Market Size by Regions: 2015 VS 2020 VS 20262.2.2 Blood and Bone Marrow Cancer Treatment Historic Market Share by Regions (2015-2020)2.2.3 Blood and Bone Marrow Cancer Treatment Forecasted Market Size by Regions (2021-2026)2.3 Industry Trends and Growth Strategy2.3.1 Market Top Trends2.3.2 Market Drivers2.3.3 Market Challenges2.3.4 Porters Five Forces Analysis2.3.5 Blood and Bone Marrow Cancer Treatment Market Growth Strategy2.3.6 Primary Interviews with Key Blood and Bone Marrow Cancer Treatment Players (Opinion Leaders) 3 Competition Landscape by Key Players3.1 Global Top Blood and Bone Marrow Cancer Treatment Players by Market Size3.1.1 Global Top Blood and Bone Marrow Cancer Treatment Players by Revenue (2015-2020)3.1.2 Global Blood and Bone Marrow Cancer Treatment Revenue Market Share by Players (2015-2020)3.1.3 Global Blood and Bone Marrow Cancer Treatment Market Share by Company Type (Tier 1, Tier 2 and Tier 3)3.2 Global Blood and Bone Marrow Cancer Treatment Market Concentration Ratio3.2.1 Global Blood and Bone Marrow Cancer Treatment Market Concentration Ratio (CR5 and HHI)3.2.2 Global Top 10 and Top 5 Companies by Blood and Bone Marrow Cancer Treatment Revenue in 20193.3 Blood and Bone Marrow Cancer Treatment Key Players Head office and Area Served3.4 Key Players Blood and Bone Marrow Cancer Treatment Product Solution and Service3.5 Date of Enter into Blood and Bone Marrow Cancer Treatment Market3.6 Mergers & Acquisitions, Expansion Plans 4 Breakdown Data by Type (2015-2026)4.1 Global Blood and Bone Marrow Cancer Treatment Historic Market Size by Type (2015-2020)4.2 Global Blood and Bone Marrow Cancer Treatment Forecasted Market Size by Type (2021-2026) 5 Blood and Bone Marrow Cancer Treatment Breakdown Data by Application (2015-2026)5.1 Global Blood and Bone Marrow Cancer Treatment Market Size by Application (2015-2020)5.2 Global Blood and Bone Marrow Cancer Treatment Forecasted Market Size by Application (2021-2026) 6 North America6.1 North America Blood and Bone Marrow Cancer Treatment Market Size (2015-2020)6.2 Blood and Bone Marrow Cancer Treatment Key Players in North America (2019-2020)6.3 North America Blood and Bone Marrow Cancer Treatment Market Size by Type (2015-2020)6.4 North America Blood and Bone Marrow Cancer Treatment Market Size by Application (2015-2020) 7 Europe7.1 Europe Blood and Bone Marrow Cancer Treatment Market Size (2015-2020)7.2 Blood and Bone Marrow Cancer Treatment Key Players in Europe (2019-2020)7.3 Europe Blood and Bone Marrow Cancer Treatment Market Size by Type (2015-2020)7.4 Europe Blood and Bone Marrow Cancer Treatment Market Size by Application (2015-2020) 8 China8.1 China Blood and Bone Marrow Cancer Treatment Market Size (2015-2020)8.2 Blood and Bone Marrow Cancer Treatment Key Players in China (2019-2020)8.3 China Blood and Bone Marrow Cancer Treatment Market Size by Type (2015-2020)8.4 China Blood and Bone Marrow Cancer Treatment Market Size by Application (2015-2020) 9 Japan9.1 Japan Blood and Bone Marrow Cancer Treatment Market Size (2015-2020)9.2 Blood and Bone Marrow Cancer Treatment Key Players in Japan (2019-2020)9.3 Japan Blood and Bone Marrow Cancer Treatment Market Size by Type (2015-2020)9.4 Japan Blood and Bone Marrow Cancer Treatment Market Size by Application (2015-2020) 10 Southeast Asia10.1 Southeast Asia Blood and Bone Marrow Cancer Treatment Market Size (2015-2020)10.2 Blood and Bone Marrow Cancer Treatment Key Players in Southeast Asia (2019-2020)10.3 Southeast Asia Blood and Bone Marrow Cancer Treatment Market Size by Type (2015-2020)10.4 Southeast Asia Blood and Bone Marrow Cancer Treatment Market Size by Application (2015-2020) 11 India11.1 India Blood and Bone Marrow Cancer Treatment Market Size (2015-2020)11.2 Blood and Bone Marrow Cancer Treatment Key Players in India (2019-2020)11.3 India Blood and Bone Marrow Cancer Treatment Market Size by Type (2015-2020)11.4 India Blood and Bone Marrow Cancer Treatment Market Size by Application (2015-2020) 12 Central & South America12.1 Central & South America Blood and Bone Marrow Cancer Treatment Market Size (2015-2020)12.2 Blood and Bone Marrow Cancer Treatment Key Players in Central & South America (2019-2020)12.3 Central & South America Blood and Bone Marrow Cancer Treatment Market Size by Type (2015-2020)12.4 Central & South America Blood and Bone Marrow Cancer Treatment Market Size by Application (2015-2020) 13 Key Players Profiles13.1 AstraZeneca, Plc.13.1.1 AstraZeneca, Plc. Company Details13.1.2 AstraZeneca, Plc. Business Overview and Its Total Revenue13.1.3 AstraZeneca, Plc. Blood and Bone Marrow Cancer Treatment Introduction13.1.4 AstraZeneca, Plc. Revenue in Blood and Bone Marrow Cancer Treatment Business (2015-2020))13.1.5 AstraZeneca, Plc. Recent Development13.2 Celgene, Inc.13.2.1 Celgene, Inc. Company Details13.2.2 Celgene, Inc. Business Overview and Its Total Revenue13.2.3 Celgene, Inc. Blood and Bone Marrow Cancer Treatment Introduction13.2.4 Celgene, Inc. Revenue in Blood and Bone Marrow Cancer Treatment Business (2015-2020)13.2.5 Celgene, Inc. Recent Development13.3 Bristol Myers Squibb & Company13.3.1 Bristol Myers Squibb & Company Company Details13.3.2 Bristol Myers Squibb & Company Business Overview and Its Total Revenue13.3.3 Bristol Myers Squibb & Company Blood and Bone Marrow Cancer Treatment Introduction13.3.4 Bristol Myers Squibb & Company Revenue in Blood and Bone Marrow Cancer Treatment Business (2015-2020)13.3.5 Bristol Myers Squibb & Company Recent Development13.4 Eli Lilly & Company13.4.1 Eli Lilly & Company Company Details13.4.2 Eli Lilly & Company Business Overview and Its Total Revenue13.4.3 Eli Lilly & Company Blood and Bone Marrow Cancer Treatment Introduction13.4.4 Eli Lilly & Company Revenue in Blood and Bone Marrow Cancer Treatment Business (2015-2020)13.4.5 Eli Lilly & Company Recent Development13.5 Johnson & Johnson Company13.5.1 Johnson & Johnson Company Company Details13.5.2 Johnson & Johnson Company Business Overview and Its Total Revenue13.5.3 Johnson & Johnson Company Blood and Bone Marrow Cancer Treatment Introduction13.5.4 Johnson & Johnson Company Revenue in Blood and Bone Marrow Cancer Treatment Business (2015-2020)13.5.5 Johnson & Johnson Company Recent Development13.6 F.Hoffman La-Roche Ltd.13.6.1 F.Hoffman La-Roche Ltd. Company Details13.6.2 F.Hoffman La-Roche Ltd. Business Overview and Its Total Revenue13.6.3 F.Hoffman La-Roche Ltd. Blood and Bone Marrow Cancer Treatment Introduction13.6.4 F.Hoffman La-Roche Ltd. Revenue in Blood and Bone Marrow Cancer Treatment Business (2015-2020)13.6.5 F.Hoffman La-Roche Ltd. Recent Development13.7 Merck & Co., Inc.13.7.1 Merck & Co., Inc. Company Details13.7.2 Merck & Co., Inc. Business Overview and Its Total Revenue13.7.3 Merck & Co., Inc. Blood and Bone Marrow Cancer Treatment Introduction13.7.4 Merck & Co., Inc. Revenue in Blood and Bone Marrow Cancer Treatment Business (2015-2020)13.7.5 Merck & Co., Inc. Recent Development13.8 Novartis AG13.8.1 Novartis AG Company Details13.8.2 Novartis AG Business Overview and Its Total Revenue13.8.3 Novartis AG Blood and Bone Marrow Cancer Treatment Introduction13.8.4 Novartis AG Revenue in Blood and Bone Marrow Cancer Treatment Business (2015-2020)13.8.5 Novartis AG Recent Development13.9 Pfizer, Inc.13.9.1 Pfizer, Inc. Company Details13.9.2 Pfizer, Inc. Business Overview and Its Total Revenue13.9.3 Pfizer, Inc. Blood and Bone Marrow Cancer Treatment Introduction13.9.4 Pfizer, Inc. Revenue in Blood and Bone Marrow Cancer Treatment Business (2015-2020)13.9.5 Pfizer, Inc. Recent Development13.10 Varian Medical Systems, Inc.13.10.1 Varian Medical Systems, Inc. Company Details13.10.2 Varian Medical Systems, Inc. Business Overview and Its Total Revenue13.10.3 Varian Medical Systems, Inc. Blood and Bone Marrow Cancer Treatment Introduction13.10.4 Varian Medical Systems, Inc. Revenue in Blood and Bone Marrow Cancer Treatment Business (2015-2020)13.10.5 Varian Medical Systems, Inc. Recent Development 14 Analysts Viewpoints/Conclusions 15 Appendix15.1 Research Methodology15.1.1 Methodology/Research Approach15.1.2 Data Source15.2 Disclaimer15.3 Author Details

About Us:

QYResearch always pursuits high product quality with the belief that quality is the soul of business. Through years of effort and supports from huge number of customer supports, QYResearch consulting group has accumulated creative design methods on many high-quality markets investigation and research team with rich experience. Today, QYResearch has become the brand of quality assurance in consulting industry.

Read more:
Blood and Bone Marrow Cancer Treatment Market Overview Industry Demand, Development and Growth Forecast Report 2026 - Cole of Duty

Ability to ‘Create’ Astrocytes Supports Their Damaging Role in MS… – Multiple Sclerosis News Today

An inflammatory environment can turn astrocytes, key supportive cells for neurons, into their killers, fostering the progression of neurodegenerative diseases like multiple sclerosis (MS), a new study shows.

This work, led by researchers at the New York Stem Cell Foundation(NYSCF), created for a first time astrocytes derived from human induced pluripotent stem cells (hIPSCs). The group then placed these cells in an inflammatory environment, and observed what happened.

Now that we can create this critical brain cell type from any individuals stem cells and capture its errant behaviors, we can better understand its role in diseases like multiple sclerosis, Parkinsons, and Alzheimers, Susan L. Solomon, the CEO of theNYSCF, said in a press release.

This will shed new light on the devastating process of neurodegeneration, pointing us towards effective treatments for this growing group of patients, Solomon added.

The study CD49f Is a Novel Marker of Functional and Reactive Human iPSC-Derived Astrocytes was published in the journal Neuron.

Astrocytes compose more than half of the cells of the central nervous system (brain and spinal cord), and work as support cells. They help to maintain brain homeostasis (stable equilibrium), provide neurons with metabolic support, enhance the connectivity of neural circuits, and control the brains blood flow.

Yet, these cells are also thought to be key players in the onset and progression of neurodegenerative diseases such as MS.

Knowledge on astrocyte biology has mostly come from animal models, namely rodents, since scientists struggle to obtain astrocytes from people.

NYSCF researchers developed a method to generate functional astrocytes that are derived from human IPSCs. (IPSCs themselves arederived from either skin or blood cells that have been reprogrammed back into a stem cell-like state, which allows for the development of an unlimited source of almost any type of human cell.)

They based their work on a previous protocol, which they developed to produce oligodendrocytes one type of cell capable of producing myelin, the protective layer covering nerve fibers and whose loss triggers MS.

Here, the researchers generated a mix of astrocytes and neurons.

They then conducted a screen to identify a surface protein that could be used to specifically purify astrocytes.

The marker CD49f was found to distinguish astrocytes from neuronal progenitors and neurons. At the genetic level, cells isolated using this marker showed activity of genes characteristic of both mature and immature astrocytes. However, when researchers looked at individual cells, they saw that CD49f was more enriched in mature astrocytes.

The hIPSCs-derived astrocytes expressing CD49f helped in neuronal growth, neural communication, provided metabolism support including glutamate uptake, and secreted molecules (called cytokines) in response to inflammation triggers.

We were excited to see that our stem-cell-derived astrocytes isolated with CD49f behaved the way typical astrocytes do: they take up glutamate, respond to inflammation, engage in phagocytosis which is like cell eating and encourage mature firing patterns and connections in neurons, said Valentina Fossati, PhD,the studys lead author.

CD49f expression was found to be specific for astrocytes in samples from both healthy and diseased human brains.

We looked at human brain tissue samples from both a healthy donor and a patient with Alzheimers disease and found that these astrocytes also expressed CD49f suggesting that this protein is a reliable indicator of astrocyte identity in both health and disease, Fossati added.

Researchers next focused on addressing the question of how astrocytes misbehave in disease.

They stimulated hIPSCs-derived cells with interleukin (IL)-1b and TNF-a, two molecules known to trigger the transition of astrocytes into a neurotoxic state (called A1 reactive astrocytes) in animal models. Cells reacted by secreting pro-inflammatory cytokines, including IL-6, IL-1 alpha, and ICAM-1.

Theseastrocytes lost their capacity to uptake (absorb) glutamate, a metabolite that is toxic to neurons. They also changed their morphology, becoming constricted instead of spreading out with long arms.

To assess whether reactive A1 astrocytes would damage neurons, the team grew neurons with stimulated and unstimulated astrocytes, or treated neurons with molecules produced by astrocytes.

Astrocytes in a reactive state were seen to decrease the electric activity of neurons and to increase their apoptosis a programmed process of cell death thats a form of suicide.

These findingsdemonstrate the specific neurotoxicity of A1 hiPSC-derivedastrocytes, the researchers wrote.

They also confirmprevious work in mice, where researchers observed that inflammation turns astrocytes neurotoxic. This work was led by Shane Liddelow, PhD, an assistant professor at the NYU Grossman School of Medicine and an author of the current study.

We observed in mice that astrocytes in inflammatory environments take on a reactive state, actually attacking neurons rather than supporting them, Liddelow said.

The latest work, the researchers concluded, showed that CD49f is a reactivity-independent,astrocyte-specific cell surface antigen that is present at allstages of astrocyte development in hiPSC-derived cultures.

Astrocytes isolated with this marker recapitulatein vitrocriticalphysiological functions, they continued, and following inflammatory stimulationbecome reactive, dysfunctional, and toxic, triggering neuronaldeath all of which opens a window for the study of their role in neurodegenerative disorders.

What we saw in the dish confirmed what Dr. Liddelow saw in mice: the neurons began to die, Fossati said. Observing this rogue astrocyte phenomenon in a human model of disease suggests that it could be happening in actual patients.

She and the others now look forward to using our new system to further explore the intricacies of astrocyte function in Alzheimers, multiple sclerosis, Parkinsons, and other diseases, in the hope it will point us toward new treatment opportunities that might slowor prevent neurodegeneration.

Patricia holds her Ph.D. in Cell Biology from University Nova de Lisboa, and has served as an author on several research projects and fellowships, as well as major grant applications for European Agencies. She also served as a PhD student research assistant in the Laboratory of Doctor David A. Fidock, Department of Microbiology & Immunology, Columbia University, New York.

Total Posts: 1,053

Patrcia holds her PhD in Medical Microbiology and Infectious Diseases from the Leiden University Medical Center in Leiden, The Netherlands. She has studied Applied Biology at Universidade do Minho and was a postdoctoral research fellow at Instituto de Medicina Molecular in Lisbon, Portugal. Her work has been focused on molecular genetic traits of infectious agents such as viruses and parasites.

Read more from the original source:
Ability to 'Create' Astrocytes Supports Their Damaging Role in MS... - Multiple Sclerosis News Today

Biomarker Advances Bring New Hope of Targeted PTCL Therapy – AJMC.com Managed Markets Network

Peripheral T-cell lymphoma (PTCL) has been a challenging nemesis for scientists, in large part because it is a widely heterogeneous disease. However, new research into biomarkers for the cancer may lead to better therapies soon.

Coauthors Erin Mulvey, MD, and Jia Ruan, MD, PhD, both of Weill Cornell Medicine, in New York, write in the Journal of Hematology & Oncologythat PTCLs have long been a treatment challenge because they are heterogenous and rare. The term PTCL represents a group of non-Hodgkin lymphomas (NHLs) derived from mature T/NK cells, though the cancers emergence and mechanism can look different from patient to patient. An estimated 5%-10% of NHL cases in Western countries are classified as PTCLs, though rates are higher in Asia and South America.

Mulvey and Ruan say most patients with PTCL are currently treated in the same manner as aggressive B-cell NHL, and most patients receive the CHOP chemotherapy regimen of cyclophosphamide, doxorubicin, vincristine, and prednisone, even though it generally is not curative. In some cases, autologous stem cell transplantation is used as a treatment, though the authors report that relapse rates are high in PTCL patients who undergo the therapy.

The situation could be changing, though, as scientists have uncovered biomarkers for PTCL subtypes. There are now more than 30 subtype classifications, though the most common classification (30% in Western countries; 20%-25% in Asia) remains the catchall not otherwise specified (NOS) category. It has no characteristic immunophenotype.

Aside from PTCL, NOS, other common subtypes include nodal T cell lymphoma with T follicular helper (TFH) including angioimmunoblastic T cell lymphoma (AITL) and systemic anaplastic large cell lymphoma (sALCL).

The authors outline a number of approaches to treating the disease. Among them is targeting surface cell receptors. For instance, they write that using the antibody-drug conjugate brentuximab vedotin to target CD30+ PTCL has boosted survival when used in combination with chemotherapy in a frontline setting.

Another approach is to target the epigenome of the cancer. Epigenetic modifying agents, such as histone deacetylase (HDAC) inhibitors and hypomethylating agents appear to be effective, and Mulvey and Ruan note that therapies are currently in development that would use combination strategies incorporating these epigenetic modifying agencies as therapies in the frontline or relapsed setting.

Meanwhile, the authors write that a number of novel agents targeting proliferative signaling pathways and the tumor microenvironment are also in development and have shown promise.

Going forward, Mulvey and Ruan say researchers are working on using chimeric antigen receptor T (CAR-T) cells against CD30, an idea thats currently in a phase 2 trial for patients with relapsing/remitting CD30+ PTCL. Another effort is an antibody, AMF13, which contains binding sites for both CD30 and for CD16A, which the authors note is the receptor associated with activation of NK cells and is also being investigated for use in patients with CD30+ lymphomas.

Lastly, the authors say, the pan-T cell antigen T cell receptor -chain (TCRB) is being investigated as a target in patients with TRBC1+ selected relapsing remitting PTCL.

In their conclusion, Mulvey and Ruan say these findings suggest a changing paradigm that will deliver significantly better results for patients with PTCL.

Delivering the most effective treatment tailored to underlying biology and therapeutic targets in the first-line and relapsed settings is poised to make the most enduring impact on [a] patients survival outcome, they wrote.

Reference

Mulvey, E., Ruan, J. Biomarker-driven management strategies for peripheral T cell lymphoma. J Hematol Oncol.2020;59(13) doi:10.1186/s13045-020-00889-z

Original post:
Biomarker Advances Bring New Hope of Targeted PTCL Therapy - AJMC.com Managed Markets Network

The American Academy of Stem Cell Physicians Will Host Its Scheduled August 1, 2020 Meeting Despite COVID 19 Crisis – Yahoo Finance

AASCP will be holding their much anticipated meeting via virtual technology

MIAMI, June 4, 2020 /PRNewswire/ --A highly anticipated and sought after AASCP meeting is back by popular demand, and topics will include a Safety StandardsPanel session as well as:Sphenopalatine Ganglion Treatment, Management of COVID-19 by PhotodynamicViral Inactivation, Caudal Block Applications, Benefits, and Indications andDiagnosis and Treatment of Extra-articular Pain in Regenerative Medicine, to name a few.

The importance of this virtual conference coincides with the ever-emerging growthof the globalregenerative medicine marketwhich is expected to reachUSD 79.8 billionby 2024, at aCAGR of 20.5%from 2018 to 2024. Factors driving the growth of the market areincreasing prevalence of degenerative and chronic diseases, technological advancements in nanotechnology, bioengineering and stem cell therapy, and increasing geriatric population across the globe.TheAASCP virtual meeting is open for registration.The meeting is set for August 1, 2020.Moderators to be announced.

Due to COVID-19, the meeting will take place virtually.Thisis an effective way to ensure that everyone that wishes to participate but cannot travel, can. It will be more easily accessible to many and more economical and students, educators and physicians will not miss out on all the important topics that AASCP has on the pipeline.Virtual Workshop Lecturers will demonstrate the techniques that they spoke on with patients brought into their office on video. These virtualinteractive workshops will feature small participant-to-instructor ratios with a customized curriculum focusing on developing hands-on skills. Each technique will be taught by experts in the field, using didactic sessions with dynamic multimedia presentations, live demonstrations and scanning on live model, as well as phantoms.Registration is now open and selling out fast.

According to AASCP, if you are using biologics in your practice, whether you are using SVF, PRP, bone marrow, UCB, amniotic products,exosomes,xenografts, or peptides, there are key considerations to take into account to achieve the best safety for your patients. Please do not miss the AASCP FDA Safety Standards panel discussion, this August 1, 2020. The panel discusses FDA safety standards in the industry and AASCP offers many recommendations. Onefor example is communication with the Chief Scientific Officer from the laboratory you work with.AASCP advises that just talking to a sales agent is not sufficient enough when determining the quality of products for your patients. Sales agents typically do not have a medical or scientific background.

The spokesman for the AASCP, Dr. AJFarshchian,said earlier: "The American Academy of Stem Cell Physicians is a group of physicians, scientists and researchers who collectively represent the most authoritativenon-federal group advocating for guidelines and education on stem cell therapy and regenerative medicine. AASCP members are experts within all fields of stem cell therapy from: SVF, BM, UCB,Exosomes, Peptides,Xenografts,Allograftsand Amniotic Fluids and are considered the most experienced leaders for proper advocacy in the field."

Dr. Farshchianexplains, "We will duplicate everything we did in our past meetings such as offer 8 CME credits, have lectures, workshops, discuss FDA safety Standards, have board examinations and a virtual graduation ceremony. AASCP is offering this virtual meeting so that nobody misses out on the education."

The American Academy of Stem Cell Physicians (AASCP) is a non-profit organization created to advance research and the development of therapeutics in regenerative medicine, including diagnosis, treatment, and prevention of disease related to or occurring within the human body. Secondarily, the AASCPaims to serve as an educational resource for physicians, scientists, and the public in diseases that can be caused by physiological dysfunction that isameliorable to medical treatment.

For further information, please contact MarieBarbaat AASCP 305-891-4686and you can also visit us at http://www.aascp.net.

Related Images

dr-farshchian-teaching-at-aaoscp.jpeg Dr. Farshchian teaching at AAOSCP workshop Meeting will now be virtual

Related Links

AASCP registration

View original content to download multimedia:http://www.prnewswire.com/news-releases/the-american-academy-of-stem-cell-physicians-will-host-its-scheduled-august-1-2020-meeting-despite-covid-19-crisis-301070934.html

SOURCE American Academy of Stem Cell Physicians

Read more from the original source:
The American Academy of Stem Cell Physicians Will Host Its Scheduled August 1, 2020 Meeting Despite COVID 19 Crisis - Yahoo Finance

Human interleukin-4treated regulatory macrophages promote epithelial wound healing and reduce colitis in a mouse model – Science Advances

Abstract

Murine alternatively activated macrophages can exert anti-inflammatory effects. We sought to determine if IL-4treated human macrophages [i.e., hM(IL4)] would promote epithelial wound repair and can serve as a cell transfer treatment for inflammatory bowel disease (IBD). Blood monocytes from healthy volunteers and patients with active and inactive IBD were converted to hM(IL4)s. IL-4 treatment of blood-derived macrophages from healthy volunteers and patients with inactive IBD resulted in a characteristic CD206+CCL18+CD14low/ phenotype (RNA-seq revealed IL-4 affected expression of 996 genes). Conditioned media from freshly generated or cryopreserved hM(IL4)s promoted epithelial wound healing in part by TGF, and reduced cytokine-driven loss of epithelial barrier function in vitro. Systemic delivery of hM(IL4) to dinitrobenzene sulphonic acid (DNBS)treated Rag1/ mice significantly reduced disease. These findings from in vitro and in vivo analyses provide proof-of-concept support for the development of autologous M(IL4) transfer as a cellular immunotherapy for IBD.

Elegant studies in mice have revealed the processes of tissue seeding and replenishment with macrophages, how the microenvironment affects macrophage function, and the spectrum of macrophage phenotype, ranging from the interferon- (IFN-)/lipopolysaccharide (LPS) classically activated through a variety of alternatively activated macrophage (AAM) subtypes; the most studied being interleukin-4 (IL-4)treated macrophages [M(IL4)] (13). While capable of producing pro-inflammatory mediators, the AAM, or regulatory macrophage (Mreg), is considered anti-inflammatory, with important roles in homeostasis and tissue restitution/recovery after injury (4, 5).

Adoptive transfer of M(IL4)s reduces the severity of colitis in murine animal models (6, 7), an observation recapitulated with other Mreg subtypes (8). Arginase and IL-10 and recruitment to the gut are implicated in the anti-colitic effect observed after systemic delivery of murine AAMs (7, 911). Therapeutic benefits of AAMs have also been shown in, for example, models of inflammatory renal disease and diabetes (12, 13). Furthermore, AAMs are capable of promoting induction or development of regulatory T cells (Tregs) (14), and one consequence of mesenchymal stem cell transfer (MSCT) is the induction of Mregs (15). A detailed awareness of the mouse macrophage has accrued, where, for example, stimuli such as IL-6 and dead neutrophils enhance an M(IL4) phenotype (16, 17); however, precise understanding of human AAMs has lagged significantly behind knowledge of their murine counterparts.

The accumulation of macrophages in the mucosa of patients with inflammatory bowel disease (IBD) led to the concept that monocytes recruited to the gut are more pathogenic than resident macrophages that perform their phagocytic duties in a nonphlogistic manner (18). Consistent with this, patients with IBD can have elevated numbers of circulating pro-inflammatory CD14+CD16+ monocytes, and apheresis of these cells has some efficacy in Crohns disease (19). In addition, examination of biopsies from inflamed regions of the colon of patients with Crohns disease revealed reduced numbers of CD68+CD206+ cells, presumptive Mregs (6).

Thus, the goals of the current study were as follows: (i) using RNA sequence analysis, to assess the impact of IL-4 on human blood-derived macrophages and determine the ability of blood-derived macrophages from healthy donors and patients with IBD to convert to M(IL4s); (ii) to assess the human (h) M(IL4)s wound healing ability in an in vitro epithelial injury model; and (iii) to determine the hM(IL4)s capacity to alleviate colitis in T cell and B celldeficient Rag1/ mice treated with dinitrobenzene sulfonic acid (DNBS).

RNA sequence analysis revealed statistically significant changes in expression of 996 genes in IL-4treated macrophages, corroborating the changes in 90 immune-related genes reported by Martinez et al. (20) (Fig. 1A): 510 genes were up-regulated and 486 down-regulated (Fig. 1A). Consistent with various reports, markers indicative of an alternatively activated M(IL4) were altered (Fig. 1B), and a panel of genes associated with immune signaling and tissue repair was up-regulated (Fig. 1B). Increased expression of CD206 and CCL18 and down-regulation of CD14 in M(IL4)s were confirmed at the mRNA and protein levels by quantitative polymerase chain reaction (qPCR) and enzyme-linked immunosorbent assay (ELISA) or flow cytometry, respectively (Fig. 1, C and D). Specificity was confirmed using IFN-, which did not evoke this response from the macrophages (fig. S1, A and B). Gene pathway analytics showed up-regulation of signaling networks related to IL-4 and IL-10 signaling, fatty acid metabolism, and degranulation (Fig. 1E). Comparison of an immune-panel gene array on murine M(IL4) with the human RNA sequence data revealed significant changes in 18 common genes, and of these, 12 showed a similar increase or decrease (Fig. 1F). hM(IL4)s were hyporesponsive to LPS as defined by tumor necrosis factor (TNF), IL-6, granulocyte-macrophage colony-stimulating factor (GM-CSF), and MCP-1 (monocyte chemoattractant protein-1) output (fig. S2).

Blood monocytes from three healthy donors were converted to macrophages (2 105) with macrophage colony-stimulating factor (M-CSF) and then left untreated [M(0)] or treated with IL-4 [M(IL4)] or IFN- [M(IFN-)] for 48 hours (both 10 ng/ml). (A) RNA sequence (RNAseq) analysis revealed IL-4evoked significant changes in 996 genes as shown in the volcano plot and confirmed some of the previously reported gene changes (Venn diagram). (B) Heat maps showing IL-4 regulation of selected genes related to M(IL4) polarization and immune function (GF, growth factors). (C) IL-4evoked increased expression of CD206 and CCL18, and decreased CD14 mRNA was confirmed by qPCR (D) and at the protein level by ELISA or flow cytometry (MFI, mean fluorescence intensity; each dot represents macrophages from an individual donor; mean SEM; *, **, and ***P < 0.05, 0.01, and 0.001 compared to M0, respectively). (E) Reactome network analysis shows clusters of gene changes increased in M(IL4). (F) Comparison of hM(IL4) RNA sequence data with murine mRNA immune array shows good alignment [, gene change is opposite direction; , no response in mouse M(IL4)].

Monolayers of Caco2 epithelia treated with hM(IL4) conditioned medium (CM) showed enhanced wound recovery compared to culture medium only, similar to that evoked by transforming growth factor (TGF) (Fig. 2, A and B) (data not shown). hM(IFN-)-CM did not enhance epithelial wound recovery (fig. S1B). Analysis of cryopreserved hM(IL4) revealed maintenance of the CD206+CCL18+CD14low/ phenotype, although expression was often less than in freshly differentiated hM(IL4) from the same individual (fig. S3A). CM from cryopreserved hM(IL4)s promoted epithelial wound repair (fig. S3B).

(A) Representative images of epithelia showing the original margin of wounds (X), leading edge of wounds (dashed line), and leading edge (le) of the control monolayer. (B) Treatment with TGF (10 ng/ml) or CM from IL-4treated macrophages [M(IL4) CM] increased epithelial repair. The M(IL4) CM was also (C) boiled or (D) treated with trypsin, which blocked repair. (E) The hM(IL4)s spontaneously produced more TGF than nonactivated macrophages (M0) from the same donor. (F) Addition of TGF-neutralizing antibodies (1D11) to the M(IL4) CM significantly impaired epithelial repair [mean SEM; n = 6 monolayers from three experiments; *, #, and P < 0.05 compared to control (culture medium only), , P < 0.01 compared to M(IL4) CM; M0 CM, and M(IL4) CM, respectively].

Boiling or trypsin treatment of the hM(IL4)-CM reduced its ability to enhance epithelial wound recovery (Fig. 2, C and D). TGF was increased in hM(IL4)-CM compared to M(0)-CM from the same individual (Fig. 2E), and immunoneutralization of TGF significantly reduced the hM(IL4)-CM capacity to promote epithelial wound recovery (Fig. 2F). Boiling the M(IL4)-CM appeared to be more effective than the immunoneutralization of TGF in reducing wound repair, and while this may represent variation between experiments, it may suggest that heat-sensitive factors other than TGF contribute to M(IL4)-CMevoked wound repair. Redundancy in wound repair in the gut would be advantageous, given the importance of gut barrier function to health.

The addition of IL-6 enhanced the M(IL4) phenotype, with the hM(IL4 + IL6) displaying increased CD206 mRNA and CCL18 production (fig. S4, A and B). hM(IL4 + IL6)-CM produced the greatest increase in epithelial monolayer repair after wounding (fig. S4C). Comparison of hM(IL4)-CM from the same donor revealed a significant increase in epithelial recovery following treatment with hM(IL4 + IL6)-CM (n = 4, P < 0.05).

IFN- decreases epithelial barrier function when applied to the basolateral side of filter-grown epithelial monolayers, as gauged by decreased transepithelial resistance (TER) and increased transcytosis of fluorescein isothiocyanate (FITC)dextran, indicators of paracellular permeability (21). This cytokine-evoked reduction in epithelial barrier function was significantly reduced by hM(IL4)-CM (Fig. 3, A and B).

Confluent, filter-grown T84 epithelial cell monolayers were treated with IFN- (10 ng/ml) 50% CM from hM(IL4)s (1 106 cultured for 24 hours), and TER was measured 24 hours later (A) (n = 17 monolayers from six experiments; ****P < 0.001 compared to control; ##P < 0.01 compared to IFN-). (B) Following 24 hours of exposure to IFN- M(IL4) CM, 70-kDa FITC-dextran was added to the lumen aspect of the monolayers, and samples from the basolateral compartment of the culture well were collected 4 hours later and assessed (n = 4 monolayers, one experiment; inset shows TER of the monolayers under the corresponding conditions; *P < 0.001 compared to control; #P < 0.01 compared to IFN-).

Macrophages differentiated from blood monocytes from patients with inactive Crohns disease or ulcerative colitis were converted to an M(IL4) that was indistinguishable from healthy donor M(IL4) based on CD206, CCL18, and CD14 mRNA expressions (Fig. 4, A to C). There was significant variability in the responses of macrophages from patients with active IBD, with results suggesting IL-4 responders and nonresponders (Fig. 4, A to C). Review of patient characteristics did not reveal any feature(s) that predicted responsiveness to IL-4. Flow cytometry of the blood-derived macrophages from healthy volunteers and patients with Crohns disease or ulcerative colitis revealed no differences in IL-4R expression (data not shown). Exposure to pro-inflammatory cytokines in the blood could render cells hyporesponsive to IL-4; macrophages from healthy volunteers were exposed to a mixture of IFN-, IL-1, and TNF for 48 hours. When challenged with IL-4, these cells still showed increases in CD206 and CCL18 mRNA (fig. S5).

Blood-derived macrophages from healthy volunteers and patients with Crohns disease (CD) or ulcerative colitic (UC) with active (CD-A) or inactive (CD-I) disease was exposed to IL-4 (10 ng/ml, 48 hours) and (A) CD206, (B) CCL18, and (C) CD14 mRNA assessed by qPCR and normalized to nontreated macrophages (M0) from the same individual. (D) Confluent monolayers of the Caco2 epithelial cell line were mechanically wounded and exposed to culture media or a 50% CM from the various macrophage groups, and the area recovered was measured 24 hours later. (E) Setting a 1.5-fold increase in CD206 mRNA in response to IL-4 predicted an M(IL4) CM that would significantly increase epithelial wound repair [mean SEM; *P < 0.05 compared to M0 (A to C) or culture medium only (D) by analysis of variance (ANOVA) followed by Tukeys test; each dot represents macrophages from a different donor]. (F) Correlation of patient mRNA expression of TGF and CD206 with their wound healing capacity (% wound healing compared to control).

Paralleling the mRNA expression analysis, M(IL4)-CM from patients with inactive Crohns disease or ulcerative colitis significantly improved repair in wounded Caco2 epithelial monolayers (Fig. 4D). While the size of repaired epithelial monolayer was on average small, this was consistent with data from healthy donor M(IL4)-CM. Addressing the variability within patient-derived M(IL4) to promote wound repair in the in vitro assay (as with mRNA responses, there appeared to be responders and nonresponders in blood monocytes from patients with active disease), CD206 and TGF mRNAs were postulated as predictive of enhanced epithelial wound healing capacity. Using CD206 expression as a canonical hM(IL4) marker, cells with 50% increase in CD206 mRNA had the ability to significantly enhance epithelial wound healing (n = 22), whereas macrophages below this threshold had a negligible capacity to promote healing (n = 10) (Fig. 4E). Adding in consideration of M(IL4) TGF mRNA returned the finding that CM from M(IL4)s with >1.5-fold increases in CD206 and TGF mRNA had the greatest ability to promote wound healing (Fig. 4F).

Intrarectal administration of DNBS to Rag1/ mice produces an acute inflammatory response as defined by macroscopic disease score, shortening of the colon, and histopathology (Fig. 5 and fig. S6). Disease was significantly less severe in mice given either freshly differentiated (Fig. 5, A to C) or cryopreserved hM(IL4)s (Fig. 5, D to F) by intraperitoneal or intravenous administration [cryopreservation of hM(IL4) resulted in 74 7% cell survival (n = 3)]. Adoptive transfer of hM(0) did not affect the outcome of DNBS-induced colitis (Fig. 5, A to C). Terminal deoxynucleotidyl transferasemediated deoxyuridine triphosphate nick end labeling (TUNEL) staining confirmed major cell death in DNBS-treated mice that was less apparent in the hM(IL4)-treated mice (Fig. 6A). Mice receiving hM(IL4)s had increased tissue levels of IL-10 (Fig. 6B) mRNA that resulted in a shift in the murine TNF:IL-10 mRNA ratio (Fig. 6C) in favor of anti-inflammatory conditions. In accordance, hM(IL4) + DNBStreated mice had reduced colonic protein levels of TNF (Fig. 6D). In contrast, murine TGF protein levels were increased in the colon hM(IL4)-treated mice (Fig. 7A). Murine epithelia are responsive to human TGF as demonstrated by increased SMAD-phosphorylation, which was also increased when M(IL4) CM was added to a murine epithelial cell line (Fig. 7B). In this acute model of colitis, the benefit of hM(IL4) was not accompanied by any obvious increase in markers of fibrosis in the colon (table S1), liver, lung, heart, or kidney (fig. S7A).

Male C57BL/6 Rag1/ mice were treated with freshly generated hM(IL4)s or murine (m) M(IL4)s or unstimulated macrophages (M0) [106 cells intraperitoneally (i.p.)] 48 hours before intrarectal delivery of 5 mg of DNBS. Seventy-two hours later, mice were necropsied and (A) a macroscopic disease score calculated, (B) colon length recorded, and (C) histopathology assessed on H&E sections. In separate studies, hM(IL4)s cryopreserved for 1 month were thawed and delivered by i.p. or intravenous (i.v.) injection 48 hours before DNBS and subsequently, (D) disease activity score, (E) colon length, and (F) histopathology were assessed [mean SEM; each dot represents a mouse; macrophages from 10 healthy donors in five experiments (A to C) and 5 to 6 donors in two experiments for cryopreserved hM(IL4) (D to F); * and #P < 0.05 compared to control (con) and DNBS, respectively, by ANOVA followed by Tukeys test (colon length) or the Kruskal-Wallis test (disease and histopathology scores)].

Male C57/bl6 Rag1/ were treated with cryopreserved hM(IL4)s (106 cells i.p. or i.v.) and 48 hours later received 5 mg of DNBS intrarectally. Seventy-two hours later, mice were necropsied and (A) cryosections of mid-colon assessed by TUNEL staining for apoptotic cells [red, 4,6-diamidino-2-phenylindole; green, TUNEL+; original objective, 20)], (B) murine TNF and IL-10 mRNA assessed by qPCR, (C) the ratio of TNF:IL-10, and protein levels of (D) murine TNF assessed by ELISA [mean SEM; each dot represents a mouse; macrophages from four to six healthy donors in two experiments; * and #P < 0.05 compared to control (con) and DNBS, respectively, by ANOVA followed by Tukeys test; m, muscle; l, lumen; arrow, TUNEL+ cell].

Fresh or cryopreserved hM(IL4) (106 cells i.p.) were administered to male C57BL/6 Rag1/ mice and 48 hours later received intrarectal delivery of 5 mg of DNBS. Three days later, mice were necropsied and (A) colonic protein levels of TGF assessed by ELISA (mean SEM; each dot represents a mouse; macrophages from four healthy donors in two experiments; * and #P < 0.05 compared to control and DNBS, respectively, by ANOVA followed by Tukeys test). (B) CM (50%) from unstimulated M(0)s and M(IL4)s, and mouse and human TGF (10 ng/ml) were applied to serum-starved (1 hour) confluent mouse IEC.4 cells, total protein was isolated at 30 min, and SMAD2 phosphorylation was detected by immunoblotting. -actin was used as a control.

Rag1/ mice injected (intraperitoneally) with hM(IL4)s 4 weeks before DNBS were protected from the pro-colitic stimulus as assessed by disease activity scores, colon length, and degree of histopathology (Fig. 8, A to D) [a similar phenomenon occurred with wild-type BALB/c mice treated with murine M(IL4)s (Fig. 8E)]. Analysis of lung, liver, and colon of the mice given hM(IL4)s showed no increase in markers of fibrosis compared to controls (fig. S7B) as gauged by assessment of Mason trichromestained sections.

Male C57BL/6 Rag1/ mice were administered cryopreserved hM(IL4)s (106 cells i.p.) and 30 days later received intrarectal delivery of 5 mg of DNBS. Seventy-two hours later, mice were necropsied and (A) a macroscopic disease score was calculated, (B) colon length was recorded, and (C) histopathology assessed on H&E sections was assessed [data are mean SEM; each dot represents a mouse; macrophages from three healthy donors in two experiments; * and #P < 0.05 compared to control (con) and DNBS, respectively, by ANOVA followed by Tukeys test (colon length) or the Kruskal-Wallis test (disease and histopathology scores)]. (D) presents representative histological images from the treatment groups (M, outer layers of muscle; L, gut lumen; *, ulceration; triangle, inflammatory infiltrate; original objective, 20). (E) shows that male BALB/c mice treated with murine M(IL4)s 2 to 21 days before DNBS are protected from the pro-colitic stimulus [mean SEM; * and #P < 0.05 compared to control (con) and DNBS, respectively, by ANOVA followed by Kruskal-Wallis test].

Fibrosis is not a major feature of acute models of colitis, but could be initiated by M(IL4)s. Treatment of a human fibroblast cell line with hM(IL4)-CM did not result in enhanced proliferation but did increase total protein levels in the cells (table S1). qPCR revealed no increase in smooth muscle actin (-SMA) or P4-hydroxylase mRNA in the treated fibroblasts (table S1). Assessment of murine colon 5 days after receiving hM(IL4)s revealed no increase in -SMA, P4-hydroxylase, and oe collagen type III 1 mRNA (table S1).

Despite advances in IBD treatment, the best therapies can induce and maintain remission in approximately one-third of patients leaving an unmet need for safe and effective therapies. The search for a single therapy that is transformative for all patients remains elusive, and given the heterogeneous nature of IBD, an individualized approach to the patient is desirable. A substantial body of evidence reveals that many subtypes of murine AAMs are anti-inflammatory and promote healing (6, 17, 22). However, while an hM(IL4) phenotype has been defined, little is known of its function, and the markers that distinguish it from a pro-inflammatory macrophage differ from those that characterize its murine counterpart (20). We present data in support of the hypothesis that autologous M(IL4) transfer can treat IBD.

Multiple markers have been used to define hM(IL4)s, and these were confirmed and others identified by RNA sequence analysis. Setting CD206high/CCL18+/CD14/low expression as indicating an hM(IL4), CM from these cells significantly improved epithelial wound healing in a reductionist in vitro model. In addition, CM from cyropreserved hM(IL4)s was almost as effective in promoting wound healing as freshly differentiated hM(IL4)s. This adds to the practicality of hM(IL4) therapy, such that patients M(IL4)s can be stored for use upon disease relapse (11). While the increased recovery of the damaged epithelium was small in magnitude, this was a consistent finding. Marginal, but significant, increases in wound recovery when translated to the surface area of the gut could benefit to the patient. The hM(IL4)-evoked wound recovery was mediated, at least partially by TGF, adding to the postulate that manipulating TGF signaling could be therapeutic in IBD (23). Aligning with this, the reduction in barrier function caused by exposure to IFN- was significantly reduced in T84 cell monolayers cotreated with hM(IL4)-CM: findings compatible with TGF antagonism of IFN-evoked increase in epithelial permeability (24). As a caveat, TGF therapy has the specter of fibrosis, and Mregs can be fibrotic in murine models. However, not all studies report a profibrotic effect of murine M(IL4)s, even with repeated administrations, (9), while others suggest an antifibrotic effect of mouse Mregs (10).

Distinct differences exist between circulating and tissue-resident immune cells from healthy individuals and people with IBD (19). A prerequisite for autologous cell transfer is that a patients blood-derived macrophages convert to M(IL4)s. Differentiation of M(IL4)s from monocytes of patients with inactive IBD was readily achieved, and the CM promoted epithelial wound healing to a degree not appreciably different from M(IL4)s from healthy individuals. In contrast, and not unexpectedly, ~50% of patients with active IBD had an impaired ability to produce M(IL4)s with wound healing capability, an inability that correlated with reduced expression of CD206 mRNA and, in some instances, TGF mRNA. Together, the latter suggests that M(IL4) CD206 and TGF expression could be a biomarker to predict responsiveness to hM(IL4)-transfer therapy.

Many mechanisms could account for monocytes from patients with active IBD not converting to M(IL4)s (e.g., reduced signal transducers and activators of transcription 6 signaling or increased expression of suppressor of cytokine-signaling proteins). Can this inability to produce M(IL4)s be overcome? From a translational perspective, the simplest solution would be to obtain monocytes from patients in remission for conversion to M(IL4) and cryopreservation. Alternatively, the potential to drive an M(IL4)-type cell via synergism with additional stimuli could be pursued. For instance, IL-6 increases the murine M(IL4) phenotype (16), and hM(IL4 + IL6) from healthy individuals displayed the greatest increase in CD206 mRNA expression, and hM(IL4 + IL6)-CM led to enhancement of wound recovery compared to hM(IL4)-CM from the same individual.

The use of human cells while instructive and yielding proof-of-principle data must be viewed within the limitations of in vitro analyses. Using mice lacking T and B cells, adoptive transfer of freshly differentiated or cryopreserved hM(IL4)s protected Rag1/ mice from DNBS-induced colitis. Aligning with the in vitro analyses, colon from protected mice had increased levels of TGF, which could be of mouse or human origin, and participated in the suppression of disease, noting that human TGF can activate murine cells (Fig. 7B). In demonstrating in vivo efficacy, this finding also reveals that the anti-colitic effect of hM(IL4) transfer is not dependent on adaptive immunity in the recipient (11). Extrapolating to IBD, this suggests that hM(IL4) transfer could be effective in patients with deficits in their T cells (25). Furthermore, the anti-colitic effect of hM(IL4)s occurred when transfer was performed 4 weeks before treatment with DNBS. A similar sustained response was reported for a murine Mreg in a mouse model of diabetes (13). Mechanistically, this implies either prolonged establishment of the cells (perhaps due to a lack of adaptive immunity in Rag1/ mice) or that the M(IL4) initiates a long-lived program in the recipient, rendering them less susceptible to DNBS; these possibilities should be assessed. In addition, we cannot exclude the possibility that death of M(IL4)s in vivo has an anti-colitic effect and should be addressed in subsequent mechanistic studies. The longevity of the hM(IL4) effect in the Rag1/-DNBS model, if translatable, presents the intriguing possibility that autologous transfer in patients could enhance recovery from a flare in disease and extend the period of remission. Experiments designed to determine the location of the transferred cells could be used to enhance hM(IL4) therapy by, for example, using cells in which mucosal (or gut specific) addressins are increased.

When proposing a new therapy, the issue of safety is omnipresent. Assessment of direct interaction between hM(IL4)s and human fibroblasts, albeit in an acute in vitro setting, revealed a small increase in fibroblast total protein, consistent with increased growth, but with negligible changes in markers of fibrosis. Also, Rag1/ mice displayed no overt fibrosis following hM(IL4) treatment. Nevertheless, fibrosis remains an unknown risk with hM(IL4) and requires further study. Susceptibility to bacterial infection is a concern (26), as with any immunosuppressive therapy (e.g., anti-TNF therapy), and while mouse M(IL4)s retain a phagocytic capacity that can be boosted by butyrate (27), we should be cognizant of this possibility with hM(IL4) transfers for IBD. Also, immunosuppressive macrophages can promote cancer (28), and studies to assess hM(IL4) transfer as a potential oncogenic stimulus need to be conducted.

Cellular immunotherapy for IBD is not unprecedented, such as MSCT for fistulizing disease (29). Intriguingly, murine and human MSCTs increase AAMs in mice and people, (15) which can, in turn, promote development of Tregs: (30) reciprocally, and Tregs can promote AAM development (31). Thus, AAMs may be at the center of a regulatory pathway with MSCs and Tregs that protects tissues from inflammatory damage. Many of the issues surrounding MSCT for IBD (e.g., sex, disease location, and concomitant therapy) (32) also apply to hM(IL4) transfer to treat IBD and need to be addressed in rigorous studies.

Having shown that IL-4 evokes major transcriptome changes in human blood monocytederived macrophages and that hM(IL4)s promote epithelial wound repair in an in vitro assay, reduce cytokine-induced epithelial barrier defects, and are beneficial in a murine model of acute colitis, we have demonstrated the cells pro-healing/anti-inflammatory ability and present this as proof-of-concept support for M(IL4) immunotherapy for IBD.

Our overall objective was to determine whether hM(IL4)s promote epithelial wound healing and suppress colitis, as proof-of-principle data to support future analysis of autologous cell transfer therapy with M(IL4)s for treating IBDs. Blood-derived macrophages were isolated from healthy donors and from patients with active and inactive Crohns disease and ulcerative colitis, and conversion to an M(IL4) phenotype was assessed via qPCR and flow cytometry; in addition, RNA-sequencing analysis was applied to M(IL4)s from three healthy donors for a broader appreciation of M(IL4) phenotype. An in vitro assay was used to determine the wound healing capacity of M(IL4)s by applying CM from these cells to mechanically wounded Caco2 epithelial monolayers for 48 hours, followed by measurement of total area repaired. To assess the protective effect of M(IL4)s in vivo, Rag1/ mice were administered fresh or cryopreserved hM(IL4)s (intraperitoneally or intravenously) 2 days or 1 month before intrarectal instillation of the pro-colitic agent, DNBS, and colitis was assessed 3 days later by macroscopic disease activity score, histopathology scores, TUNEL staining, and qPCR and ELISA for IL-10, TGF, and TNF. Scoring of disease was performed in a blinded fashion. The processing of samples and statistical analysis was executed simultaneously for all experimental groups. For all experiments, a minimum of three experiments and a minimum n value of four were chosen. The amount of replicates and statistics are referred to in each figure legend.

Animal experiments adhered to the Canadian Council on Animal Welfare as administered by the University of Calgary Animal Care Committee under protocol AC17-0115. Male 10- to 14-week-old C57BL/6 Rag1/ mice (breeding colony, University of Calgary) were injected intraperitoneally or intravenously with 106 hM(IL4) or hM(0) in 250 l of phosphate-buffered saline 48 hours before intrarectal delivery of 2,4-DNBS (5 mg in 100 l of 50% ethanol; MP Biomedicals) to induce colitis (11). In another set of experiments, mice received hM(IL4) 1 month before DNBS treatment. Murine M(IL4) served as a positive control (11).

On day 3 after DNBS, necropsies were performed, and disease was assessed by a macroscopic disease activity score. A segment of mid-colon was excised, fixed, and processed for H&E staining, and a histopathology score was calculated. Collagen deposition was examined in Massons trichromestained sections of formalin-fixed colon, liver, heart, lung, and kidney (6, 9). In some experiments, colonic and liver levels of collagen were measured using the Sircol Soluble Collagen Assay (Biocolor assays). Additional portions of colon were excised and extracted for total protein to determine TNF and TGF levels by ELISA or placed in TRIzol (Invitrogen) and TNF and IL-10 mRNA assessed by qPCR.

Blood from healthy volunteers [male = 15, female = 9; age range, 20 to 66, mean = 31 12 (SD)] was obtained with approval by the University of Calgary Conjoint Health Research Ethics Board (REB15-1270_REN3). Patients [table S2; male = 17, female = 16; age range, 20 to 87, mean = 47 17 (SD)] were consented, and blood was obtained through the University of Calgary Intestinal Inflammation Tissue bank (REB15-0586_REN3). Peripheral blood mononuclear cells were collected, and following a 4-hour 37C incubation on petri dishes, nonadherent cells were removed, and the adherent cells were incubated in culture medium (RPMI-1640 medium), 2% penicillin/streptomycin, Hepes (20 mM; all from Gibco), and 10% pooled normal human heat-inactivated and sterile-filtered AB serum (Innovative Research, Novi, MI) with recombinant hM-CSF (10 ng/ml) for 7 days. Medium was changed on day 5, with the addition of hM-CSF. Size, granularity, and CD68 expression by flow cytometry revealed that the cultures were 97.5 0.1% macrophages (n = 3). Macrophages (2.5 105) were differentiated for 48 hours with recombinant IL-4 (IL-6) or IFN- (all 10 ng/ml) and then assessed. Differentiated macrophages (2.5 105) received 1 ml of fresh culture medium, and 24 hours later, the CM was collected for use and cytokine determinations. Differentiated macrophages are not a rapidly dividing cell type; however, we assessed cell numbers 72 hours and 1 week after cytokine treatment and found no significant change in cell numbers [i.e., proliferation; M(0) = 2.6 0.7 compared to M(IL4) = 2.7 0.07 105] and % cell survival [i.e., viability; M(0) = 88 16 compared to M(IL4) = 95 26], respectively (n = 4; mean SD). Similarly, testing of viability via trypan blue exclusion revealed no differences between the two groups at the end of the in vitro culture when either CM was collected or cells harvested for delivery to mice.

For cryopreservation, M(IL4)s were resuspended (5 105/ml) in 10% dimethyl sulfoxide in human serum and stored at 80C overnight in Mr. Frosty containers (Thermo Fisher Scientific, Wilmington, DE) with isopropanol and then transferred to liquid nitrogen.

Macrophage RNA was isolated using the Aurum Total RNA Mini Kit (Bio-Rad Laboratories, Hercules, CA), quantified using the Nanodrop 1000 Spectrophotometer (Thermo Fisher Scientific), and 0.5 g of RNA converted to complementary DNA (cDNA) using an iScript kit (Bio-Rad Laboratories Canada, Mississauga, ON, Canada). Real-time qPCR of human and murine macrophages was performed (9) using primers listed in table S3. Data are presented as the target gene: 18S ribosomal RNA gene ratio for M(0) that was normalized to a value for 1 for comparison with M(IL4) from the same individual. A correlation graph of the expression of human TGF mRNA, CD206 mRNA, and % epithelial wound healing from M(IL4)-CM was made in R (version 3.5.3) using the ggplot2 (version 3.2.1).

RNA from M(0) and M(IL4)s from three healthy donors was quality assessed using the Agilent Tapestation 2200, with RNA integrity values 8 to 10 indicating samples free of DNA contamination (21). RNA sequencing was performed at the Centre for Health Genomics and Informatics, University of Calgary, where samples were made into libraries using the Illumina TruSeq Stranded mRNA LT library preparation kit and were sequenced on a 75-cycle high-output NextSeq 500 run. Briefly, transcripts were quantified with Kallisto 0.43.1 (33) using Homo sapiens GRCh38 (Ensembl release 90) cDNA, with sequence bias correction turned on and 50 bootstraps. All downstream analyses were done in R 3.5.1 (34). Sleuth 0.30.0 was used for differential expression, with treatment as the main effect and donor included as a covariate (35). The M(IL4)s were compared to nonstimulated M(0) using the Wald test, and differentially expressed genes were selected on the basis of a q value cutoff of 0.05. Enriched gene sets for both KEGG (36) and Reactome (37) pathways were identified with overrepresentation tests (P < 0.05) using clusterProfiler 3.10.0 (38) and ReactomePA 1.26.0 (39), respectively. Data were deposited in NCBIs Gene Expression Omnibus and are accessible through GEO Series accession number GSE132732 (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE132732).

Human colonderived Caco2 or T84 epithelial cells were maintained in Dulbeccos modified Eagles medium (DMEM) [supplemented with 10% fetal bovine serum (FBS), 2% penicillin/streptomycin, and 0.00144% sodium bicarbonate]. Cells (5 105) were seeded in six-well plates for 48 hours and transferred to DMEM (0.5% FBS) for 24 hours, and monolayers were wounded with a razor blade. Macrophage CM was diluted 1:1 with DMEM culture medium (0.5% human serum) and applied to wounded epithelia for 48 hours. Wounds were observed using the Nikon Eclipse Ti-U inverted microscope at 4 magnification (Melville, NY) (40). Analysis of repair was measured in three fields of view from the initial wound edge to the leading edge for total area repaired in micrometer2. When CM from patient cells was used, wound repair was normalized for % baseline increase of total area repaired compared to medium control. TGF (10 ng/ml) served as a positive control. In some experiments, CM was (i) boiled (20 min) and (ii) subjected to trypsin (2 U/l; 37C; 2 hours) and then trypsin inhibitors (2 U/l for 2 ml of CM; 37C; 30 min), or (iii) TGF-neutralizing antibodies were added (100 ng/ml; 2 hours at 37C; 1d11 clone Cedarlane Labs).

T84 epithelial cells were seeded onto 3-m porous filter supports and cultured until confluent (24) as determined by a TER of 1000 ohm.cm2. Monolayers were then treated basolaterally with IFN- (10 ng/ml) 50% M(IL4)-CM, and TER was measured 24 hours later via voltmeter and chopstick electrodes. Then, 70-kDa FITC-dextran (200 g/ml; Sigma Chemical Co.) was added to the apical side of the monolayer, samples (500 l) of basolateral medium were collected 4 hours later, fluorescence was measured, and amount of FITC-dextran was read off a standard curve.

Cell culture medium was collected from nonactivated M(0) or M(IL4) or LPS (10 ng/ml for 24 hours)activated macrophages and was subjected to the Luminex human cytokine array pro-inflammatory focused 13-plex (HDF13) panel (EveTechnologies, Calgary, Canada). ELISA DuoSet kits (R&D Systems Inc.) were used to measure CCL18, IL-10, TGF, and TNF in macrophage supernatants according to the manufacturers protocols. All samples were assessed in duplicate.

Confluent mouse IEC4.1 intestinal epithelial cells were serum starved for 1 hour and then treated with CM from hM(0) and hM(IL4)s and mouse or human recombinant TGF (10 ng/ml, 30 min; eBioscience). Whole extracts were analyzed by immunoblotting protocol with the following antibodies: rabbit polyclonal phospho-SMAD2 (1:1000, Cell Signaling Technology, #3108), rabbit polyclonal SMAD2 (1:2500, Cell Signaling Technology, #3102), and rabbit polyclonal -actin (1, 1000, Abcam, #8227).

The human colonic fibroblast cell line CCD-18Co (American Type Culture Collection, CRL-1459) was seeded (5 105) in six-well plates for 48 hours and then treated with M(0)-CM or M(IL4)-CM (50%), and mRNA, total protein, and proliferation were assessed.

Parametric data were analyzed using one-way analysis of variance (ANOVA) with Tukeys posttest or by Students t test for two group comparisons. When data were normalized, analysis was one-way ANOVA with Kruskal-Wallis posttest or Wilcoxon signed-rank test. Statistical comparisons were performed using GraphPad Prism 5.0 (GraphPad Prism Software, La Jolla, CA), and a level of statistically significant difference was accepted at P < 0.05.

This is an open-access article distributed under the terms of the Creative Commons Attribution-NonCommercial license, which permits use, distribution, and reproduction in any medium, so long as the resultant use is not for commercial advantage and provided the original work is properly cited.

Acknowledgments: T.S.J. was supported, in part, by a stipend from the NSERC CREATE Host-Parasite Interactions (HPI) program at the University of Calgary. G.L. held an AI-HS PhD studentship. A.S. was supported by the Beverly Philips Snyder Institute University of Calgary, NSERC CREATE HPI, and the Canadian Association of Gastroenterology studentships. D.M.M. holds a Canada Research Chair (Tier 1) in Intestinal Immunophysiology in Health and Disease. We acknowledge support from the Intestinal Tissue Bank and G. Bindra at the University of Calgary for access to patient material and the flow cytometry suite (University of Calgary). Funding: Funding for this study was provided by a Crohns Colitis Canada Grant-in-Aid to D.M.M. Author contributions: T.S.J.: study concept and design, acquisition of data, analysis and interpretation of data, drafting of the manuscript, and statistical analysis; G.L.: acquisition of data and analysis and interpretation of data; A.W.: acquisition of data; B.E.C.: acquisition of data; M.L.W.: analysis and interpretation of data, statistical analysis, and technical support; S.R.: acquisition of data; A.S.: acquisition of data; N.M.: acquisition of data; P.L.B.: study concept and design and critical revision of manuscript for important intellectual content; R.P.: study concept and design and critical revision of manuscript for important intellectual content; D.M.M.: study concept and design, interpretation of data, drafting of the manuscript, critical revision of the manuscript for important intellectual content, obtained funding, and study supervision. Competing interests: All authors declare that they have no competing interests. Data and materials availability: All data needed to evaluate the conclusions in the paper are present in the paper and/or the Supplementary Materials. Additional data related to this paper may be requested from the authors.

Read more from the original source:
Human interleukin-4treated regulatory macrophages promote epithelial wound healing and reduce colitis in a mouse model - Science Advances

Stem Cell Banking Market to Witness Huge Growth in Coming Years 2020-2027 – Jewish Life News

Stem cell banking or preservation is a combined process of extraction, processing and storage of stem cells, so that they may be used for treatment of various medical conditions in the future, when required. Stem cells have the amazing power to get transformed into any tissue or organ in the body. In recent days, stem cells are used to treat variety of life-threatening diseases such as blood and bone marrow diseases, blood cancers, and immune disorders among others.

The market of stem cell banking is anticipated to grow with a significant rate in the coming years, owing to factors such as, development of novel technologies for stem cell preservation and processing, and storage; growing awareness on the potential of stem cells for various therapeutic conditions. Moreover, increasing investments in stem cell research is also expected to propel the growth of the stem cell banking market across the globe. On other hand rising burden of major diseases and emerging economies are expected to offer significant growth opportunities for the players operating in stem cell banking market.

Get Sample PDF Copy athttps://www.theinsightpartners.com/sample/TIPBT00002082/

Key Points of the Report:

In addition, the report focuses on leading industry players with information such as company profiles, components and services offered, financial information of last 3 years, key development in past five years.

1. Cordlife2. ViaCord (A Subsidiary of PerkinElmer)3. Cryo-Save AG4. StemCyte India Therapeutics Pvt. Ltd.5. Cryo-Cell International, Inc.6. SMART CELLS PLUS.7. Vita 348. LifeCell9. Global Cord Blood Corporation10. CBR Systems, Inc.

The Insight Partners has segmented the global Stem Cell Banking Market:

The source segment includes, placental stem cells (PSCS), dental pulp-derived stem cells (DPSCS), bone marrow-derived stem cells (BMSCS), adipose tissue-derived stem cells (ADSCS), human embryo-derived stem cells (HESCS), and other stem cell sources. Based on service type the market is segmented into, sample processing, sample analysis, sample preservation and storage, sample collection and transportation. Based on application, the market is segmented as, clinical applications, research applications, and personalized banking applications.

The report covers key developments in the Stem Cell Banking market as organic and inorganic growth strategies. Various companies are focusing on organic growth strategies such as product launches, product approvals and others such as patents and events. Inorganic growth strategies witnessed in the market were acquisitions, and partnership & collaborations. These activities have paved way for expansion of business and customer base of market players.

The report provides a detailed overview of the industry including both qualitative and quantitative information. It provides overview and forecast of the global Stem Cell Banking market based on various segments. It also provides market size and forecast estimates from year 2017 to 2027 with respect to five major regions, namely; North America, Europe, Asia-Pacific (APAC), Middle East and Africa (MEA) and South & Central America. The Stem Cell Banking market by each region is later sub-segmented by respective countries and segments. The report covers analysis and forecast of 18 countries globally along with current trend and opportunities prevailing in the region.

The report analyzes factors affecting Stem Cell Banking market from both demand and supply side and further evaluates market dynamics affecting the market during the forecast period i.e., drivers, restraints, opportunities, and future trends. The report also provides exhaustive PEST analysis for all five regions namely; North America, Europe, APAC, MEA and South & Central America after evaluating political, economic, social and technological factors effecting the Stem Cell Banking market in these regions.

Purchase this Report at:https://www.theinsightpartners.com/buy/TIPBT00002082/

About Us:The Insight Partners is a one stop industry research provider of actionable intelligence. We help our clients in getting solutions to their research requirements through our syndicated and consulting research services. We are a specialist in Technology, Semiconductors, Healthcare, Manufacturing, Automotive and Defense.

Contact Us:

Contact Person: Sameer JoshiPhone: +1-646-491-9876Email Id:[emailprotected]heinsightpartners.com

The rest is here:
Stem Cell Banking Market to Witness Huge Growth in Coming Years 2020-2027 - Jewish Life News

The bioreactors market was valued at US$ 2,958.50 million in 2019 and is projected to reach US$ 5,169.01 million by 2027; it is expected to grow at a…

New York, June 05, 2020 (GLOBE NEWSWIRE) -- Reportlinker.com announces the release of the report "Bioreactors Market Forecast to 2027 - COVID-19 Impact and Global Analysis by Cell ; Molecule ; Technology ; End User, and Geography" - https://www.reportlinker.com/p05908638/?utm_source=GNW

A bioreactor is equipment or system engineered to provide biologically active environment for the production of various medical and pharmaceutical compounds.The vessel is used to carry out a chemical process, which involves organisms or biochemically active substances derived from such organisms.

This process can either be aerobic or anaerobic.The bioreactors are commonly cylindrical, ranging in size from liters to cubic meters, and are generally made of stainless steel.

Bioreactors provide a controllable environment, in terms of pH, temperature, nutrient supply, and shear stress for any cells.The use of single-use bioreactors has increased in the modern biopharmaceutical processes in the last few years.This can be attributed to their unique ability to allow enhanced process flexibility, reduce investment requirements, and limit operational costs.

Also, many companies have developed single-use bioreactors for the production of a wide range of therapeutics. For instance, Distek Inc., has developed a benchtop single-use bioreactor system for recombinant protein production. Single-use bioreactors reduce the risks of contamination and decrease production turnaround times. Moreover, the reduction in validation time has been one of the prime benefits of single-use bioreactors. The rising adoption of single-use bioreactors for upstream bioprocessing is driving the growth of the market. For instance, Sartorius AG offers a wide range of single-use bioreactors. The company provides ambr 15 for a 10-15 mL microbioreactor scale and Biostat STR for 50-2000 L.The global bioreactors market is segmented on the basis of cell, molecule, technology, and end user.The bioreactors market, by molecule, is segmented into monoclonal antibodies, vaccines, recombinant proteins, stem cells, gene therapy, and others.

The monoclonal antibodies segment held the largest share of the market in 2019.However, the stem cell segment is projected to register the highest CAGR in the market during the forecast period.

Based on cell, the bioreactors market is segmented into mammalian cells, bacterial cells, yeast cells, and others.Based on technology, the market is segmented into wave-induced motion sub, stirred sub, single-use bubble column, and others.

Based on end user, the market is segmented into research and development organizations, biopharma manufacturers, contract manufacturing organizations (CMOs).A few of the essential primary and secondary sources referred to while preparing the report are the Food and Drug Administration (FDA), World Health Organization (WHO), Organization for Economic Co-operation and Development, National Institutes of Health (NIH), and Centers for Disease Control and Prevention (CDC), among others.Bioreactors Market Forecast to 2027 - Covid-19 Impact and Global Analysis by Cell, Molecule, Technology, End User, and GeographyThe bioreactors market was valued at US$ 2,958.50 million in 2019 and is projected to reach US$ 5,169.01 million by 2027; it is expected to grow at a CAGR of 7.3% from 2020 to 2027. A bioreactor is equipment or system engineered to provide biologically active environment for the production of various medical and pharmaceutical compounds.The vessel is used to carry out a chemical process, which involves organisms or biochemically active substances derived from such organisms.

This process can either be aerobic or anaerobic.The bioreactors are commonly cylindrical, ranging in size from liters to cubic meters, and are generally made of stainless steel.

Bioreactors provide a controllable environment, in terms of pH, temperature, nutrient supply, and shear stress for any cells.The use of single-use bioreactors has increased in the modern biopharmaceutical processes in the last few years.This can be attributed to their unique ability to allow enhanced process flexibility, reduce investment requirements, and limit operational costs.

Also, many companies have developed single-use bioreactors for the production of a wide range of therapeutics. For instance, Distek Inc., has developed a benchtop single-use bioreactor system for recombinant protein production. Single-use bioreactors reduce the risks of contamination and decrease production turnaround times. Moreover, the reduction in validation time has been one of the prime benefits of single-use bioreactors. The rising adoption of single-use bioreactors for upstream bioprocessing is driving the growth of the market. For instance, Sartorius AG offers a wide range of single-use bioreactors. The company provides ambr 15 for a 10-15 mL microbioreactor scale and Biostat STR for 50-2000 L.The global bioreactors market is segmented on the basis of cell, molecule, technology, and end user.The bioreactors market, by molecule, is segmented into monoclonal antibodies, vaccines, recombinant proteins, stem cells, gene therapy, and others.

The monoclonal antibodies segment held the largest share of the market in 2019.However, the stem cell segment is projected to register the highest CAGR in the market during the forecast period.

Based on cell, the bioreactors market is segmented into mammalian cells, bacterial cells, yeast cells, and others.Based on technology, the market is segmented into wave-induced motion sub, stirred sub, single-use bubble column, and others.

Based on end user, the market is segmented into research and development organizations, biopharma manufacturers, contract manufacturing organizations (CMOs).A few of the essential primary and secondary sources referred to while preparing the report are the Food and Drug Administration (FDA), World Health Organization (WHO), Organization for Economic Co-operation and Development, National Institutes of Health (NIH), and Centers for Disease Control and Prevention (CDC), among others.Read the full report: https://www.reportlinker.com/p05908638/?utm_source=GNW

About ReportlinkerReportLinker 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.

__________________________

Original post:
The bioreactors market was valued at US$ 2,958.50 million in 2019 and is projected to reach US$ 5,169.01 million by 2027; it is expected to grow at a...