Page 21234..»

Category Archives: Iowa Stem Cells

Fate Therapeutics Inc. (FATE) and KemPharm Inc. (NASDAQ:KMPH) Comparison side by side – The EN Herald

Posted: September 23, 2019 at 6:46 am

This is therefore a contrasting of the risk, analyst recommendations, institutional ownership, profitability, dividends, earnings and valuation in Fate Therapeutics Inc. (NASDAQ:FATE) and KemPharm Inc. (NASDAQ:KMPH). The two are both Biotechnology companies that compete with one another.

Valuation and Earnings

Table 1 shows top-line revenue, earnings per share (EPS) and valuation of the two companies.

Profitability

Table 2 represents Fate Therapeutics Inc. (NASDAQ:FATE) and KemPharm Inc. (NASDAQ:KMPH)s net margins, return on equity and return on assets.

Volatility & Risk

A 1.62 beta indicates that Fate Therapeutics Inc. is 62.00% more volatile compared to S&P 500. Competitively, KemPharm Inc.s beta is 1.33 which is 33.00% more volatile than S&P 500.

Liquidity

Fate Therapeutics Inc.s Current Ratio is 6.4 while its Quick Ratio is 6.4. On the competitive side is, KemPharm Inc. which has a 1 Current Ratio and a 1 Quick Ratio. Fate Therapeutics Inc. is better positioned to pay off short and long-term obligations compared to KemPharm Inc.

Analyst Ratings

The next table highlights the delivered recommendations and ratings for Fate Therapeutics Inc. and KemPharm Inc.

Fate Therapeutics Inc. has a 30.49% upside potential and a consensus price target of $23.75. Meanwhile, KemPharm Inc.s average price target is $1.05, while its potential upside is 28.83%. Based on the analysts view we can conclude, Fate Therapeutics Inc. is looking more favorable than KemPharm Inc.

Institutional and Insider Ownership

Institutional investors owned 98.8% of Fate Therapeutics Inc. shares and 33.5% of KemPharm Inc. shares. Fate Therapeutics Inc.s share owned by insiders are 0.9%. Comparatively, KemPharm Inc. has 0.5% of its share owned by insiders.

Performance

In this table we show the Weekly, Monthly, Quarterly, Half Yearly, Yearly and YTD Performance of both pretenders.

For the past year Fate Therapeutics Inc. had bullish trend while KemPharm Inc. had bearish trend.

Summary

On 8 of the 9 factors Fate Therapeutics Inc. beats KemPharm Inc.

Fate Therapeutics, Inc., a clinical-stage biopharmaceutical company, develops programmed cellular immunotherapies for cancer and immune disorders worldwide. Its immuno-oncology product candidates include FATE-NK100, a natural killer (NK) cell cancer immunotherapy that consists of adaptive memory NK cells; engineered hnCD16 induced pluripotent stem cells (iPSC)-derived natural killer cell therapy candidates for hematologic/solid tumors; and engineered chimeric antigen receptor iPSC-derived T cell therapy product candidates for hematologic/solid tumors. The companys immuno-regulation product candidates comprise ProTmune, an investigational programmed cellular immunotherapy for use as a next-generation allogeneic hematopoietic cell transplantation cell graft; and ToleraCyte for the treatment of autoimmune and inflammatory diseases; engineered iPSC-derived CD34+ cell therapy for immune disorders. Fate Therapeutics, Inc. has a research collaboration and license agreement with Memorial Sloan Kettering Cancer Center to develop off-the-shelf T-cell immunotherapies; and strategic research collaboration and license agreement with Juno Therapeutics, Inc. to identify small molecule modulators that enhance the function of T cells. Fate Therapeutics, Inc. was founded in 2007 and is headquartered in San Diego, California.

KemPharm, Inc., a clinical-stage specialty pharmaceutical company, discovers and develops new proprietary prodrugs in the United States. Its lead product candidates are KP415, an extended release d-threo-methylphenidate product candidate for the treatment of ADHD; and KP201/IR, an IR formulation of KP201, a prodrug of hydrocodone and acetaminophen for the treatment of acute pain. The company is also involved in developing KP511/ER, a prodrug of hydromorphone for the management of pain; KP511/IR for the short duration management of acute pain; KP606/IR, an IR formulation of KP606, a prodrug of oxycodone for the management of moderate to severe pain; KP746, a prodrug of oxymorphone for the management of moderate to severe pain; and KP303, a prodrug of quetiapine for the treatment of central nervous system disorders. KemPharm, Inc. was founded in 2006 and is headquartered in Coralville, Iowa.

Receive News & Ratings Via Email - Enter your email address below to receive a concise daily summary of the latest news and analysts' ratings with our FREE daily email newsletter.

Here is the original post:
Fate Therapeutics Inc. (FATE) and KemPharm Inc. (NASDAQ:KMPH) Comparison side by side - The EN Herald

Posted in Iowa Stem Cells | Comments Off on Fate Therapeutics Inc. (FATE) and KemPharm Inc. (NASDAQ:KMPH) Comparison side by side – The EN Herald

Common Prostate Drug May Slow Progression of Parkinson, Researchers Say – AJMC.com Managed Markets Network

Posted: September 23, 2019 at 6:46 am

Terazosin, a drug used to treat enlarged prostate, may also be able to slow the progression of Parkinson disease.

The finding is the result of a collaboration involving researchers in China and at the University of Iowa (UI), combining observations from animal experiments with information from clinical databases regarding men taking the drug.

Lei Liu, PhD, at Capital Medical University in Beijing, China, found that terazosin could block cell death. Using toxin-induced and genetic PD models in mice, rats, flies, and induced pluripotent stem cells, the drug increased brain adenosine triphosphate levels and slowed or prevented neuron loss if it was given before the onset of cell death. In addition, the drug could slow or stop neurodegeneration, even if treatment was delayed until after neurodegeneration had started to develop. Liu's team discovered that the cell-protective activity was due to terazosin's ability to activate phosphoglycerate kinase 1 (PGK1), an enzyme critical for cellular energy production.

Researchers then probed databases looking at patients who took terazosin and found slower disease progression, decreased PD-related complications, and a reduced frequency of PD diagnoses.

This suggests that in patients taking terazosin and related drugs, enhanced PGK1 activity and increased glycolysis may slow neurodegeneration in PD.

"When we tested the drug in various different animal models of PD, they all got better. Both the molecular changes in the brain associated with cell death and the motor coordination in the animals improved," said Liu, a professor in the Beijing Institute for Brain Disorders, in a statement.

Nandakumar Narayanan, MD, PhD, a UI neurologist, and Jordan Schultz, PharmD, UI assistant professor of psychiatry, examined the Parkinson's Progression Markers Initiative database, which is sponsored by The Michael J. Fox Foundation for Parkinson's Research. The data showed that men with PD who were taking terazosin had reduced rates of progressive motor disability compared to men with PD who were taking a different drug, tamsulosin, for enlarged prostate.

While tamsulosin is also used to treat benign prostatic hyperplasia, unlike terazosin, it does not have any effect on the PGK1 enzyme, making ita good control.

Only 13 men were identified who were taking terazosin or 1 of 2 similar drugs that also activate the PGK1 enzyme, compared with 293 men with PD who were either taking tamsulosin or were not taking any of these drugs. While the differences in motor decline between the 2 groups were statistically significant, the team looked to confirm the findings using the larger IBM Watson/Truven Health Analytics MarketScan Database, which includes de-identified records of more than 250 million people.

From there, researchers identified 2880 Parkinson's patients taking 1 of the 3 drugs that target PGK1 (terazosin, doxazosin, or alfusin) and a comparison group of 15,409 PD patients taking tamsulosin. Using medical codes to track PD-related diagnoses and hospital or clinic visits for all the patients, the data suggested that under real world conditions, terazosin and related drugs reduce the signs, symptoms, and complications of PD. Relative to patients with PD taking tamsulosin, those on terazosin or the 2 other drugs had reduced clinic and hospital visits for motor symptoms (relative risk [RR] 0.77; 95% CI, 0.700.84), nonmotor symptoms (RR 0.78; 95% CI, 0.730.83), and PD complications (RR 0.76; 95% CI, 0.710.82).

Patients using terazosin also had a reduced risk of a PD diagnosis, the researchers said.

Reference

Cai R, Zhang Y, Simmering JE, et al. Enhancing glycolysis attenuates Parkinsons disease progression in models and clinical databases [published online September 16, 2019].J Clin Invest. doi: 10.1172/JCI129987.

Continued here:
Common Prostate Drug May Slow Progression of Parkinson, Researchers Say - AJMC.com Managed Markets Network

Posted in Iowa Stem Cells | Comments Off on Common Prostate Drug May Slow Progression of Parkinson, Researchers Say – AJMC.com Managed Markets Network

Contrasting of Cellular Biomedicine Group Inc. (CBMG) and KemPharm Inc. (NASDAQ:KMPH) – The EN Herald

Posted: September 23, 2019 at 6:46 am

Cellular Biomedicine Group Inc. (NASDAQ:CBMG) and KemPharm Inc. (NASDAQ:KMPH), are influenced by compare since they are both players in the Biotechnology. These factors are particularly influence the analyst recommendations, profitability, risk, dividends, earnings and valuation, institutional ownership of the two firms.

Valuation and Earnings

Table 1 highlights Cellular Biomedicine Group Inc. and KemPharm Inc.s top-line revenue, earnings per share (EPS) and valuation.

Profitability

Table 2 demonstrates the return on assets, return on equity and net margins of Cellular Biomedicine Group Inc. and KemPharm Inc.

Risk and Volatility

Cellular Biomedicine Group Inc.s 2.67 beta indicates that its volatility is 167.00% more volatile than that of Standard & Poors 500. KemPharm Inc.s 33.00% more volatile than Standard & Poors 500 volatility due to the companys 1.33 beta.

Liquidity

The Current Ratio and a Quick Ratio of Cellular Biomedicine Group Inc. are 4.4 and 4.4. Competitively, KemPharm Inc. has 1 and 1 for Current and Quick Ratio. Cellular Biomedicine Group Inc.s better ability to pay short and long-term obligations than KemPharm Inc.

Analyst Ratings

The Ratings and Recommendations for Cellular Biomedicine Group Inc. and KemPharm Inc. are featured in the next table.

Cellular Biomedicine Group Inc.s consensus price target is $23, while its potential upside is 60.39%. Competitively KemPharm Inc. has an average price target of $1.05, with potential upside of 28.83%. Based on the analysts belief we can conclude, Cellular Biomedicine Group Inc. is looking more favorable than KemPharm Inc.

Institutional and Insider Ownership

Cellular Biomedicine Group Inc. and KemPharm Inc. has shares held by institutional investors as follows: 23.8% and 33.5%. 37.14% are Cellular Biomedicine Group Inc.s share held by insiders. On the other hand, insiders held about 0.5% of KemPharm Inc.s shares.

Performance

Here are the Weekly, Monthly, Quarterly, Half Yearly, Yearly and YTD Performance of both pretenders.

For the past year Cellular Biomedicine Group Inc.s stock price has smaller decline than KemPharm Inc.

Summary

Cellular Biomedicine Group Inc. beats KemPharm Inc. on 8 of the 9 factors.

Cellular Biomedicine Group Inc., a biopharmaceutical company, develops treatments for cancerous and degenerative diseases in Greater China. It focuses on developing and marketing cell-based therapies to treat serious diseases, such as cancer, orthopedic, and various inflammatory diseases, as well as metabolic diseases. The company develops treatments utilizing proprietary cell based technologies, including immune cell therapy for the treatment of a range of cancers; human adipose-derived mesenchymal progenitor cells for the treatment of joint and autoimmune diseases; and tumor cell specific dendritic cell therapy. The company has a strategic research collaboration with GE Healthcare Life Sciences China to co-develop industrial control processes in Chimeric Antigen Receptor T-cell (CAR-T) and stem cell manufacturing. Cellular Biomedicine Group Inc. was incorporated in 2001 and is headquartered in Cupertino, California.

KemPharm, Inc., a clinical-stage specialty pharmaceutical company, discovers and develops new proprietary prodrugs in the United States. Its lead product candidates are KP415, an extended release d-threo-methylphenidate product candidate for the treatment of ADHD; and KP201/IR, an IR formulation of KP201, a prodrug of hydrocodone and acetaminophen for the treatment of acute pain. The company is also involved in developing KP511/ER, a prodrug of hydromorphone for the management of pain; KP511/IR for the short duration management of acute pain; KP606/IR, an IR formulation of KP606, a prodrug of oxycodone for the management of moderate to severe pain; KP746, a prodrug of oxymorphone for the management of moderate to severe pain; and KP303, a prodrug of quetiapine for the treatment of central nervous system disorders. KemPharm, Inc. was founded in 2006 and is headquartered in Coralville, Iowa.

Receive News & Ratings Via Email - Enter your email address below to receive a concise daily summary of the latest news and analysts' ratings with our FREE daily email newsletter.

Continued here:
Contrasting of Cellular Biomedicine Group Inc. (CBMG) and KemPharm Inc. (NASDAQ:KMPH) - The EN Herald

Posted in Iowa Stem Cells | Comments Off on Contrasting of Cellular Biomedicine Group Inc. (CBMG) and KemPharm Inc. (NASDAQ:KMPH) – The EN Herald

Our Team – Neural Stem Cell Institute, Rensselaer NY

Posted: April 12, 2019 at 11:51 pm

Our Team

Working together to discover cures. Dr. Sally Temple

Christina Allen-Calabrese, Ph.D. Research Scientist

Dr. Christina Allen-Calabrese obtained a Ph.D. from the University at Albany School of Public Health in partnership with the New York State Department of Health in Biomedical Science with formal training in neuroscience. Dr. Calabrese is trained in mouse and human neural stem cell research. She also has extensive experience in Biosafety Level 2 and 3 Laboratories and GLP training. Dr. Calabrese currently studies the role of the meninges in providing specific factors to different regions of the brain and is exploring the hypothesis that perturbations in this system could contribute to neurodegenerative diseases, such as Alzheimers and Parkinsons disease.

Nathan Boles, Ph.D. Principal Investigator

Ph.D. Interdepartmental Program in Cell and Molecular Biology, Baylor College of Medicine.

Dr. Boles carried out his thesis work with Dr. Margaret A. Goodell studying the regulation of the hematopoietic stem cell. His work at NSCI explores the role of epigenetics in neural stem cell self-renewal and differentiation.More about Nathan.

Sue Borden, B.A. Research Technician

BA Biology: Russell Sage College, Biotechnology Certificate: HVCC

Sue has worked in neurobiology research labs at SUNYA, AMC and Cornell University, in clinical labs in Ithaca and the Albany area and as a teaching assistant in a local K-12 school. She recently joined the Retinal Stem Cell Consortium team as a research technician. Sue will be culturing and analyzing hRPE stem cells in preparation for clinical trials in the treatment of AMD.

David Butler, Ph.D. Principal Investigator

Dr. Butlers long-term goal is to develop novel intracellular antibody (intrabody) therapeutics forneurodegenerative disorders caused by misfolded proteins. He hasa broad background in degenerative diseases associated with aging. As a postdoc in the Messer lab, whichpioneeredthe use of intracellular antibodies in the brain, David developed bifunctional intra-cellular antibodies. Davids bifunctional antibodies were able to prevent mutant Huntingtin and Synuclein from misfolding while directing them to the proteasome for degradation. He is currently utilizing induced pluripotent stem cell (iPSC) disease modeling to develop novel bifunctional intrabody reagents for tauopathies such as FTD and Alzheimers disease. Dr. Butler is also an Adjunct Professor in the Biomedical Sciences Department, School of Public Health, SUNY Albany.More about David.

Carol Charniga, B.S. Research Technician/Manager of Operations

Carol worked in cancer research labs in Pennsylvania and at Albany Medical Center before joining AMCs Dept. of Neurosurgery and Neuroscience and the lab of Dr. Sally Temple. Since the creation of the Neural Stem Cell Institute, Carol has been involved in all projects developing CNS, spinal cord and eye as well as being the safety officer and Lab Manager of Operations. Currently, most of her workday is spent in the eye group lab, involved in the macular degeneration program.

Rebecca Chowdhury, Ph.D. Post Doctoral Fellow

Dr. Chowdhury obtained her Ph.D. from Iowa State University, studying intrinsic factors that control cell fate decisions in the developing retina. She is currently studying the role of Stau2,an RNA-binding protein (RBP) involved in neuronal development andmaturation. She isusing human induced pluripotent stem cell-derived cortical neurons and a genetically modified mouse model in her studies.More about Rebecca.

Liz Fisher, Ph.D. Post Doctoral Fellow

Dr. Fisherobtained her Ph.D. from the University of Texas Health Science Center at San Antonio, UT, studying the role of astrocyte glutamate metabolism following stroke. She then boldly made the decision to leave the warmth of south Texas for upstate New York in October 2017. As a post-doctoral fellow at the Neural Stem Cell Institute,Lizcontinues to study the roles of non-neuronal cells following injury. She is currently investigating how modulating immune cell populations using biodegradable microbeads can influence recovery following spinal cord injury.More about Liz.

Susan K. Goderie, A.A.S., B.S. Research Technician/ Manager of Research

After attending Hudson Valley Community College and Plattsburg State University, Susan worked for the New York State Birth Defects Institute culturing lymphocytes for karyology. Later she worked in the kidney transplantation lab at the Albany Medical Center. She worked with Dr. Harold Kimelberg studying astrocytic swelling in response to ischemic conditions until 1995 when she joined the lab of Dr. Sally Temple. She oversees the daily lab research; training of new faculty, staff, and students; supervising the technical staff as well as directly contributing to the spinal cord injury, stem cell niche, embryonic brain development, and macular degeneration projects.

Shona Joy, Ph.D. Post Doctoral Fellow

Dr. Joy holds two masters degree in Stem Cell Technology and Biotechnology and a Ph.D. in Stem Cells Neuroscience fromCardiff University, UK. She is interested in using pluripotent stem cells to generate models for neurodegenerative diseases(Alzheimers disease and Progressive supranuclear palsy) to facilitate regenerative medicine and drug discovery. She uses the stable induced pluripotent stem cell lines she generates to investigate the signaling pathways involved in reprogramming these cells.More about Shona.

Thomas Kiehl, Ph.D. Principal Investigator

Dr. Kiehl started his career with an M.S. in Computer Science from Rensselaer Polytechnic Institute. After 11 years at GE Global Research, in their Computational Intelligence Lab, Tom returned to RPI full time to pursue a Ph.D. in Multidisciplinary Science with a focus on systems biology and biotechnology. This was followed by a postdoc at Albany Medical College in Immunology. A Computing Innovation Fellowship, awarded by the Computing Research Association, allowed Tom to spend two years at the SUNY College of Nanoscale Science and Engineering where he began work in computational neuroscience and RNA-seq analysis. At NSCI Dr. Kiehl facilitates the integration of data analysis with bench work. Tom is also pursuing applications of high-throughput in-vitro electrophysiological platforms for the study of development, spinal cord injury, and neurological disease mechanisms. Learn more about the role of Computing@NSCI.More about Tom.

Steven Lotz, B.S.Research Technician

After college, Steve worked for Taconics Surgical Modifications Department. In 2001, he began a career as a Research Technician at the Albany Medical Center. Three years later, he joined AMCs Immunology Core as the FACS operator. In 2009 he began working at NeuraCell Bank, part of the Neural Stem Cell Institute, as the Sr. Flow Cytometry Applications Specialist.

Natalia Lowry, M.D., Ph.D. Principal Investigator

Dr. Lowry received her MD from Russian State Medical University and her Ph.D. from Albany Medical College in 2000.Dr. Lowry has been trained in mouse neural stem cell research during a post-doctoral fellowship under Dr. Sally Temple, and then joined NSCI in 2007 as aprincipalinvestigator with interest in using neural stem cells as a therapeutic tool to treat spinal cord injuries and other neurodegenerative diseases. Currently, Dr. Lowry combines her research work at NSCI with a clinical education position at

Anne Messer, Ph.D. Principal Investigator

Anne Messer, Ph.D., is a senior scientist focused on the development of novel therapeutics for degenerative diseases caused by misfolded proteins that trigger breakdowns in the functions of critical cells. She pioneered the use of engineered antibody technologies for Huntingtons and Parkinsons disease. Her recent studies range from antibodyengineering and nanobody selection to brain delivery using gene therapies. This biotechnology to harness immune processes now is being combined with stem cell studies and expanded to cover a range of important age-related diseases, including Age-relatedMacular Degeneration. More about Anne.

Khadijah Onanuga, Ph.D., PMP Director of Research Programs

Dr. Onanuga received her Ph.D.in Nanoscale Engineering with a specialty in Nanobiotechnology from the SUNY Polytechnic Institute, Colleges of Nanoscale Science and Engineering. As the Director of Research Programs at NSCI, Dr. Onanuga oversees the research programs at the institute which include: the Age-Related Macular Degeneration Program of theRetinal Stem Cell Consortium, its IND application process for a cell-based therapy, the Stem Cell Group of the Tau Consortium- a group dedicated to using stem cell technology for research and drug discovery that targets neurodegenerative diseases, and other programs.

Natasha Rugenstein, A.S.Research Technician

Tashaattended Hudson Valley Community College, NY, receiving an A.S. degree in biological sciences and the Biotechnology Certificate. She joined the Neural Stem Cell Institute in 2017, and currently works on the histology of study samples in on-going projects while continuing her education in biology.

Jeffrey Stern, M.D., Ph.D. Principal Investigator/ Director of Translational Research/ Co-Founder

Dr. Stern was trained as a biophysicist in vision research at Brandeis University, MA and Rockefeller University, NY, receiving his Ph.D. in 1982. He then studied medicine at the University of Miami Medical School and completed his residency in Ophthalmology at the Albany Medical Center. Dr. Stern did a fellowship in vitreo-retinal specialty at Mt. Sinai Medical School, NYC.More about Dr. Stern.

Sally Temple, Ph.D. Scientific Director/ Principal Investigator/ Co-Founder

Dr. Sally Temple is the co-Founder and Scientific Director of the Neural Stem Cell Institute located in Rensselaer, NY. A native of York, England, Dr. Temple leads a team of 30 researchers focused on using neural stem cells to develop therapies for eye, brain, and spinal cord disorders. In 2008, she was awarded the MacArthur Fellowship Award for her contribution and future potential in the neural stem cell field. As the Scientific Director of NSCI, Dr. Temple oversees the research mission from basic to translational projects. She is also responsible for the staff, budget, and developing the overall strategic plan for the institute. Dr. Temple is a member of the board of directors of the International Society for Stem Cell Research and of the medical advisory boards of the NY Stem Cell Foundation and the Genetics Policy Institute. Her numerous articles have been published in such journals asNature,Cell Stem Cell,Neuron,andCell.More about Sally.

Brian Unruh, B.S. Research Technician

Brian graduated from Binghamton University in 2017 and joined the NSCI shortly thereafter. His work focuses chiefly on the production, characterization, and purification of iPSC derived retinal pigment epithelial cells, as well as age-related macular degeneration disease modeling. Brian aspires to attend medical school in the nearest future.

Jenny Yue Wang, M.D. Research Technician

Jenny obtained an MD in China and worked in the University of California before joining Dr. Sally Temples lab. Her research interest and experiences include but are not limited to neural stem cell fate choice, cell culture and in vivo experiments on mice.

Xiuli Zhao, M.D., Ph.D. Post Doctoral Fellow

Dr. Zhao earned her MD from the Anhui University of Chinese Medicine and completed her training as an ophthalmologist in the first affiliated hospital of Jinan University, China. She received her Ph.D. in Neuroscience from Arizona State University in 2017. Her current research involves live cell imaging to compare neural stem cell (NSC) activity changes in the subventricular zone between young and aged mice. She is also working to identify the choroid plexus-secreted environment factors that alter mouse and human NSC activities with aging.More about Xiuli.

Cindy Butler Executive Assistant

Cindy is the go toperson at our organization. She has many years of experience handling the administrative tasks associated with running a research laboratory. Perhaps it is her previous experience in childcare that enables her to remain pleasant in even the most difficult situations.

Jake Parks Bookkeeper

Jake is responsible for the recording of data transactions into the financial accounting system and retaining the documentation for those records. Jake also supports the IT department as a first responder on the help desk and assists with the maintenance of the Institutes computer network.

Tom Irwin Administrative Director

Tom received his MBA from Bernard Baruch College City University of New York. He has worked in the academic medical environment for 30 plus years mostly in research administration at Cornell Medical College, NYC and Albany Medical College, Albany, NY. He has also served as an administrative reviewer for the NIH, IACUC institutional official, has been an institutional biosafety committee member and is currently an ex-officio member of the RPI Institutional Stem Cell Research Oversight committee (ISCRO).

The rest is here:
Our Team - Neural Stem Cell Institute, Rensselaer NY

Posted in Iowa Stem Cells | Comments Off on Our Team – Neural Stem Cell Institute, Rensselaer NY

Cultured meat – Wikipedia

Posted: January 20, 2019 at 11:45 pm

Cultured meat is meat produced by in vitro cultivation of animal cells, instead of from slaughtered animals.[1] It is a form of cellular agriculture.

Cultured meat is produced using many of the same tissue engineering techniques traditionally used in regenerative medicine.[2] The concept of cultured meat was popularized by Jason Matheny in the early 2000s after co-authoring a seminal paper[3] on cultured meat production and creating New Harvest, the world's first non-profit organization dedicated to supporting in vitro meat research.[4]

In 2013, Mark Post, professor at Maastricht University, was the first to showcase a proof-of-concept for in-vitro lab grown meat by creating the first lab-grown burger patty. Since then, several cultured meat prototypes have gained media attention: however, because of limited dedicated research activities, cultured meat has not yet been commercialized.[5] In addition, it has yet to be seen whether consumers will accept cultured meat as meat.[6]

The production process still has much room for improvement, but it has advanced in most recent years, leading up to 2018, under various companies.[7] Its applications lead it to have several prospective health, environmental, cultural, and economic considerations in comparison to conventional meat.[8]

Besides cultured meat, the terms in vitro meat, vat-grown[9], lab-grown meat,[10] cell-based meat,[11] clean meat, and synthetic meat[12] have all been used by various outlets to describe the product.

Clean meat is an alternative term that is preferred by some journalists, advocates, and organizations that support the technology. According to the Good Food Institute,[13] the name better reflects the production and benefits of the meat[14][15] and surpassed "cultured" and "in vitro" in media mentions as well as Google searches.[16]

The theoretical possibility of growing meat in an industrial setting has long captured the public imagination. Winston Churchill suggested in 1931: "We shall escape the absurdity of growing a whole chicken in order to eat the breast or wing, by growing these parts separately under a suitable medium."[17]

In vitro cultivation of muscular fibers was performed as early as 1971 by Russell Ross. Indeed, the abstract was

Smooth muscle derived from the inner media and intima of immature guinea pig aorta were grown for up to 8 weeks in cell culture. The cells maintained the morphology of smooth muscle at all phases of their growth in culture. After growing to confluency, they grew in multiple overlapping layers. By week 4 in culture, microfibrils (110 A) appeared within the spaces between the layers of cells. Basement membrane-like material also appeared adjacent to the cells. Analysis of the microfibrils showed that they have an amino acid composition similar to that of the microfibrillar protein of the intact elastic fiber. These investigations coupled with the radioautographic observations of the ability of aortic smooth muscle to synthesize and secrete extracellular proteins demonstrate that this cell is a connective tissue synthetic cell.[18]

The culturing of stem cells from animals has been possible since the 1990s, including the production of small quantities of tissue which could, in principle be cooked and eaten. NASA has been conducting experiments since 2001, producing cultured meat from turkey cells.[19][20] The first edible sample was produced by the NSR/Touro Applied BioScience Research Consortium in 2002: goldfish cells grown to resemble fish fillets.[21][22][23]

In 1998 Jon F. Vein of the United States filed for, and ultimately secured, a patent (US 6,835,390 B1) for the production of tissue engineered meat for human consumption, wherein muscle and fat cells would be grown in an integrated fashion to create food products such as beef, poultry and fish.

In 2001, dermatologist Wiete Westerhof from the University of Amsterdam, medical doctor Willem van Eelen, and businessman Willem van Kooten announced that they had filed for a worldwide patent on a process to produce cultured meat.[24] In the process, a matrix of collagen is seeded with muscle cells, which are then bathed in a nutritious solution and induced to divide.[25] Scientists in Amsterdam study the culture medium, while the University of Utrecht studies the proliferation of muscle cells, and the Eindhoven University of Technology is researching bioreactors.[25][dead link]

In 2003, Oron Catts and Ionat Zurr of the Tissue Culture and Art Project and Harvard Medical School exhibited in Nantes a "steak" a few centimetres wide, grown from frog stem cells, which was cooked and eaten.[26]

The first peer-reviewed journal article published on the subject of laboratory-grown meat appeared in a 2005 issue of Tissue Engineering.[27]

In 2008, PETA offered a $1 million prize to the first company to bring lab-grown chicken meat to consumers by 2012.[28] The Dutch government has put US$4 million into experiments regarding cultured meat.[19] The In Vitro Meat Consortium, a group formed by international researchers interested in the technology, held the first international conference on the production of cultured meat, hosted by the Food Research Institute of Norway in April 2008, to discuss commercial possibilities.[21]Time magazine declared cultured meat production to be one of the 50 breakthrough ideas of 2009.[29]In November 2009, scientists from the Netherlands announced they had managed to grow meat in the laboratory using the cells from a live pig.[30]

As of 2012, 30 laboratories from around the world have announced that they are working on cultured meat research.[31]

The first cultured beef burger patty, created by Dr. Mark Post at Maastricht University, was eaten at a demonstration for the press in London in August 2013.[32] It was made from over 20,000 thin strands of muscle tissue.[33] This burger cost Dr. Post over $300,000 to make and over 2 years to produce.[34] Two other companies have also begun to culture meat; Memphis Meats in the US and SuperMeat in Israel.[35]

As of February 2017, a recent report has shown that the price of these cultured burgers has dropped dramatically. Going from roughly over $300,000 to $11.36 in just 3 and a half years.[36] This cost is now only 9-10 times more expensive per pound than standard ground beef.[36]

On August 5, 2013, the world's first lab-grown burger was cooked and eaten at a news conference in London. Scientists from Maastricht University in the Netherlands, led by professor Mark Post, had taken stem cells from a cow and grown them into strips of muscle which they then combined to make a burger. The burger was cooked by chef Richard McGeown of Couch's Great House Restaurant, Polperro, Cornwall, and tasted by critics Hanni Rtzler, a food researcher from the Future Food Studio and Josh Schonwald.[32] Rtzler stated,

There is really a bite to it, there is quite some flavour with the browning. I know there is no fat in it so I didn't really know how juicy it would be, but there is quite some intense taste; it's close to meat, it's not that juicy, but the consistency is perfect. This is meat to me... It's really something to bite on and I think the look is quite similar.[37]

Rtzler added that even in a blind trial she would have taken the product for meat rather than a soya copy.[37]

Tissue for the London demonstration was cultivated in May 2013, using about 20,000 thin strips of cultured muscle tissue. Funding of around 250,000 came from an anonymous donor later revealed to be Sergey Brin.[38] Post remarked that "there's no reason why it can't be cheaper...If we can reduce the global herd a millionfold, then I'm happy".[39]

It's just a matter of time before this is gonna happen, I'm absolutely convinced of that. In our case, I estimate the time to be about 3years before we are ready to enter the market on a small scale, about 5years to enter the market on a larger scale, and if you'd ask me: "When will [cultured meat] be in the supermarket around the corner?" That'll be closer to 10 than to 5years, I think.

Peter Verstrate, Mosa Meat (2018)[40](1:06:15)

Since the first public trial, several startups have made advances in the field. Mosa Meat co-founded by Mark Post continuous research with a focus on cultured beef. The company was able to significantly lower the costs of production.[41]

Memphis Meats, a Silicon Valley startup founded by a cardiologist, launched a video in February 2016 showcasing its cultured beef meatball.[41][42][43] In March 2017, it showcased chicken tenders and duck a l'orange, the first cultured poultry-based foods shown to the public.[44][45][46]

An Israeli company, SuperMeat, ran a viral crowdfunding campaign in 2016 for its work on cultured chicken.[47][48][49][50][51][52]

Finless Foods, a San Francisco-based company aimed at cultured fish, was founded in June 2016. In March 2017 it commenced laboratory operations and progressed quickly. Director Mike Selden said in July 2017 to expect bringing cultured fish products on the market within two years (by the end of 2019).[53]

In March 2018, JUST, Inc. (in 2011 founded as Hampton Creek in San Francisco) claimed to be able to present a consumer product from cultured meat by the end of 2018. According to CEO Josh Tetrick the technology is already there, and now it is merely a matter of applying it. JUST has about 130 employees and a research department of 55 scientists, where lab meat from poultry, pork and beef is being developed. They would have already solved the problem of feeding the stemcells with only plant resources. JUST receives sponsoring from Chinese billionaire Li Ka-shing, Yahoo! cofounder Jerry Yang and according to Tetrick also from Heineken International amongst others.[54]

The Dutch startup Meatable, consisting of Krijn de Nood, Daan Luining, Ruud Out, Roger Pederson, Mark Kotter and Gordana Apic among others, reported in September 2018 it had succeeded in growing meat using pluripotent stem cells from animals' umbilical cords. Although such cells are reportedly difficult to work with, Meatable claimed to be able to direct them to behave using their proprietary technique in order to become muscle cells or fat cells as needed. The major advantage is that this technique bypasses fetal bovine serum, meaning that no animal has to be killed in order to produce meat.[55] That month, it was estimated there were about 30 cultured meat startups across the world. A Dutch House of Representatives Commission meeting discussed the importance and necessity of governmental support for researching, developing and introducing cultured meat in society, speaking to representatives of three universities, three startups and four civil interest groups on 26 September 2018.[40]

There are three stages in the production of cultured meat: selection of starter cells, treatment of growth medium, and scaffolding.[56][57][7]

The initial stage of growing cultured meat is to collect cells that have a rapid rate of proliferation (high cell reproduction rate). Such cells include embryonic stem cells, adult stem cells, myosatellite cells, or myoblasts. Stem cells proliferate the quickest, but have not yet begun development towards a specific kind of cell, which creates the challenge of splitting the cells and directing them to grow a certain way. Fully developed muscle cells are ideal in the aspect that they have already finished development as a muscle, but proliferate hardly at all. Therefore, cells such as myosattelite and myoblast cells are often used as they still proliferate at an acceptable rate, but also sufficiently differentiate from other types of cells.[3]

The cells are then treated by applying a protein that promotes tissue growth, which is known as a growth medium. These mediums should contain the necessary nutrients and appropriate quantity of growth factors. They are then placed in a culture medium, in a bio-reactor, which is able to supply the cells with the energetic requirements they need.[7]

To culture three-dimensional meat, the cells are grown on a scaffold, which is a component that directs its structure and order. The ideal scaffold is edible so the meat does not have to be removed, and periodically moves to stretch the developing muscle, thereby simulating the animal body during normal development. Additionally the scaffold must maintain flexibility in order to not detach from the developing myotubes (early muscle fibers). Scaffold must also allow vascularization (creation of blood vessels) in order for normal development of muscle tissue.[7][58]

Scaffold-based production techniques can only be appropriately used in boneless or ground meats (processed). The end result of this process would be meats such as hamburgers or sausages. In order to create more structured meats, for example steak, muscle tissue must be structured in directed and self-organized means or by proliferation of muscle tissue already existing. Additionally, the presence of gravitational, magnetic, fluid flow, and mechanical fields have an effect on the proliferation rates of the muscle cells. Processes of tension such as stretching and relaxing increased differentiation into muscle cells.[58]

Once this process has been started, it would be theoretically possible to continue producing meat indefinitely without introducing new cells from a living organism.[59] It has been claimed that, conditions being ideal, two months of cultured meat production could deliver up to 50,000 tons of meat from ten pork muscle cells.[60]

Cultured meat production requires a preservative, such as sodium benzoate, to protect the growing meat from yeast and fungus. Collagen powder, xanthan gum, mannitol and cochineal could be used in different ways during the process.[61]

The price of cultured meat at retail outlets like grocery stores and supermarkets may decrease to levels that middle-class consumers consider to be "inexpensive" due to technological advancements.[62][bettersourceneeded]

The science for cultured meat is an outgrowth of the field of biotechnology known as tissue engineering.[63] The technology is simultaneously being developed along with other uses for tissue engineering such as helping those with muscular dystrophy and, similarly, growing transplant organs.[19][64] There are several obstacles to overcome if it has any chance of succeeding; at the moment, the most notable ones are scale and cost.[19][21]

Additionally, there is no dedicated scientific research discipline for cellular agriculture and its development. The past research undertaken into cellular agriculture were isolated from each other, and they did not receive significant academic interest. Although it currently exists, long-term strategies are not sufficiently funded for development and severely lack a sufficient amount of researchers.[8]

Large-scale production of cultured meat may or may not require artificial growth hormones to be added to the culture for meat production.[63][70]

Researchers have suggested that omega-3 fatty acids could be added to cultured meat as a health bonus.[19] In a similar way, the omega-3 fatty acid content of conventional meat can also be increased by altering what the animals are fed.[71] An issue of Time magazine has suggested that the cell-cultured process may also decrease exposure of the meat to bacteria and disease.[21]

Due to the strictly controlled and predictable environment, cultured meat production has been compared to vertical farming, and some of its proponents have predicted that it will have similar benefits in terms of reducing exposure to dangerous chemicals like pesticides and fungicides, severe injuries, and wildlife.[72]

Concern in regards to developing antibiotic resistance due to the use of antibiotics in livestock, livestock and livestock-derived meat serving as a major source of disease outbreaks (including bird flu, anthrax, swine flu, and listeriosis), and long-term processed meat consumption beingassociated with increased heart disease, digestive tract cancer, and type 2 diabetes currently plague livestock-based meat. In regards to cultured meat, strict environmental controls and tissue monitoring can prevent infection ofmeat cultures from the outset, and any potential infection can be detected before shipment to consumers.[73]

In addition to the prevention and lack of diseases, and lack of the use of antibiotics or any other chemical substances, cultured meat can also leverage numerous biotechnology advancements, including increased nutrient fortification, individually-customized cellular and molecular compositions, and optimal nutritional profiles,all making it much healthier than livestock-sourced meat.[73]

Although cultured meat consists of genuine animal muscle cells that are the same as in traditional meat, consumers may find such a high-tech approach to food production distasteful (see appeal to nature). Cultured meat has been disparagingly described as 'Frankenmeat'.[74]

If cultured meat turns out to be different in appearance, taste, smell, texture, or other factors, it may not be commercially competitive with conventionally produced meat. The lack of fat and bone may also be a disadvantage, for these parts make appreciable culinary contributions. However, the lack of bones and/or fat may make many traditional meat preparations, such as buffalo wings, more palatable to small children.[75]

Research has suggested that environmental impacts of cultured meat would be significantly lower than normally slaughtered beef.[76] For every hectare that is used for vertical farming and/or cultured meat manufacturing, anywhere between 10 and 20 hectares of land may be converted from conventional agriculture usage back into its natural state.[77] Vertical farms (in addition to cultured meat facilities) could exploit methane digesters to generate a small portion of its own electrical needs. Methane digesters could be built on site to transform the organic waste generated at the facility into biogas which is generally composed of 65% methane along with other gasses. This biogas could then be burned to generate electricity for the greenhouse or a series of bioreactors.[78]

A study by researchers at Oxford and the University of Amsterdam found that cultured meat was "potentially ... much more efficient and environmentally-friendly", generating only 4% greenhouse gas emissions, reducing the energy needs of meat generation by up to 45%, and requiring only 2% of the land that the global meat/livestock industry does.[79][80] The patent holder Willem van Eelen,[25] the journalist Brendan I. Koerner,[81] and Hanna Tuomisto, a PhD student from Oxford University all believe it has less environmental impact.[82] This is in contrast to cattle farming, "responsible for 18% of greenhouse gases"[83] and causing more damage to the environment than the combined effects of the world's transportation system. Vertical farming may completely eliminate the need to create extra farmland in rural areas along with cultured meat.[84] Their combined role may create a sustainable solution for a cleaner environment.[84]

One skeptic is Margaret Mellon of the Union of Concerned Scientists, who speculates that the energy and fossil fuel requirements of large-scale cultured meat production may be more environmentally destructive than producing food off the land.[28] However, S.L. Davis has speculated that both vertical farming in urban areas and the activity of cultured meat facilities may cause relatively little harm to the species of wildlife that live around the facilities.[85] Dickson Despommier speculated that natural resources may be spared from depletion due to vertical farming and cultured meat, making them ideal technologies for an overpopulated world.[86] Conventional farming, on the other hand, kills ten wildlife animals per hectare each year.[85] Converting 4 hectares (10 acres) of farmland from its man-made condition back into either pristine wilderness or grasslands would save approximately 40 animals while converting 1 hectare (2 acres) of that same farmland back into the state it was in prior to settlement by human beings would save approximately 80 animals.

Additionally, the cattle industry uses a large amount of water for producing animal feed, animal rearing, and for sanitation purposes. It is estimated that the water recycled from livestock manure is contributing "33% of global nitrogen and phosphorus pollution," "50% of antibiotic pollution," "37% of toxic heavy metals," and "37% of pesticides" which contaminate the planet's freshwater.[87]

Techniques of genetic engineering, such as insertion, deletion, silencing, activation, or mutation of a gene, are not required to produce cultured meat. Furthermore, cultured meat is composed of a tissue or collection of tissues, not an organism. Therefore, it is not a genetically modified organism (GMO). Since cultured meats are simply cells grown in a controlled, artificial environment, some have commented that cultured meat more closely resembles hydroponic vegetables, rather than GMO vegetables.[88]

More research is being done on cultured meat, and although the production of cultured meat does not require techniques of genetic engineering, there is discussion among researchers about utilizing such techniques to improve the quality and sustainability of cultured meat. Fortifying cultured meat with nutrients such as beneficial fatty acids is one improvement that can be facilitated through genetic modification. The same improvement can be made without genetic modification, by manipulating the conditions of the culture medium.[89] Genetic modification may also play a role in the proliferation of muscle cells. The introduction of myogenic regulatory factors, growth factors, or other gene products into muscle cells may increase production past the capacity of conventional meat.[89]

To avoid the use of any animal products, the use of photosynthetic algae and cyanobacteria has been proposed to produce the main ingredients for the culture media, as opposed to the very commonly used fetal bovine or horse serum.[90] Some researchers suggest that the ability of algae and cyanobacteria to produce ingredients for culture media can be improved with certain technologies, most likely not excluding genetic engineering.[91]

The Australian bioethicist Julian Savulescu said "Artificial meat stops cruelty to animals, is better for the environment, could be safer and more efficient, and even healthier. We have a moral obligation to support this kind of research. It gets the ethical two thumbs up."[92]Animal welfare groups are generally in favor of the production of cultured meat because it does not have a nervous system and therefore cannot feel pain.[28][59][64] Reactions of vegetarians to cultured meat vary:[93] some feel the cultured meat presented to the public in August 2013 was not vegetarian as fetal calf serum was used in the growth medium.[94] However, since then lab grown meat has been grown under a medium that doesn't involve fetal serum.[95]

Independent inquiries may be set up by certain governments to create a degree of standards for cultured meat.[96] Laws and regulations on the proper creation of cultured meat products would have to be modernized to adapt to this newer food product.[96] Some societies may decide to block the creation of cultured meat for the "good of the people" making its legality in certain countries a questionable matter.[96]

Cultured meat needs technically sophisticated production methods making it harder for communities to produce food self-sufficiently and potentially increasing dependence on global food corporations.[97]

Independent inquiries may be set up by certain governments to create a degree of standards for cultured meat.[98] Once cultured meat becomes more cost-efficient, it is necessary to decide who will regulate the safety and standardization of these products. Prior to being available for sale, the European Union and Canada will require approved novel food applications. Additionally, the European Union requires that cultured animal products and production must prove safety, by an approved company application, which became effective as of January 1, 2018.[99] Within the United States, there is discussion of whether or not cultured meat regulation will be handled by the FDA (Food and Drug Administration) or the USDA (United States Department of Agriculture). The main point of content is whether or not cultured meat is labeled as "food" and regulated by the FDA or as a "meat food product" and regulated by the USDA. Under the FDA, cultured meat would need to follow the FFDCA and have a Food Safety Plan (FSP). Under the USDA, cultured meat would need be regulated by the FSIS who must deem the ingredients safe and usable. It could also be regulated by both government organizations.[100]

Jewish rabbinical authorities disagree whether cultured meat is kosher (food that may be consumed, according to Jewish dietary laws). However, most rabbis agree that if the original cells were taken from a kosher animal then the cultured meat will be kosher. Some even think that it would be kosher even if coming from non-kosher animals like pigs, however some disagree.[9] Some Muslim scholars have stated that cultured meat would be allowed by Islamic law if the original cells and growth medium were halal.[101] Within Hindu culture, there is significant importance of cattle in religion where the majority of Hindus reject consumption of a cow's meat. The potential of a "meatless beef" has driven debate among Hindus on the acceptance of eating it. A significant number of Hindus reject the meat due to the high prevalence of a vegetarian diet.[102]

The production of cultured meat is currently very expensive in 2008 it was about US$1 million for a piece of beef weighing 250 grams (0.55lb)[21] and it would take considerable investment to switch to large-scale production. However, the In Vitro Meat Consortium has estimated that with improvements to current technology there could be considerable reductions in the cost of cultured meat. They estimate that it could be produced for 3500/tonne (US$5424/tonne in March 2008),[103][104] which is about twice the cost of unsubsidized conventional European chicken production.[22][103]

In a March 2015 interview with Australia's ABC, Mark Post said that the marginal cost of his team's original 250,000 burger was now 8.00. He estimates that technological advancements would allow the product to be cost-competitive to traditionally sourced beef in approximately ten years.[105] In 2016, the cost of production of cultured beef for food technology company Memphis Meats was $18,000 per pound ($40,000/kg).[106] As of June 2017 Memphis Meats reduced the cost of production to below $2,400 per pound ($5,280/kg).[107]

Cultured meat has often featured in science fiction. The earliest mention may be in Two Planets (1897) by Kurd Lasswitz, where "synthetic meat" is one of the varieties of synthetic food introduced on Earth by Martians. Other notable books mentioning artificial meat include Ashes, Ashes (1943) by Ren Barjavel; The Space Merchants (1952) by Frederik Pohl and C.M. Kornbluth; The Restaurant at the End of the Universe (1980) by Douglas Adams; Le Transperceneige (Snowpiercer) (1982) by Jacques Lob and Jean-Marc Rochette; Neuromancer (1984) by William Gibson; Oryx and Crake (2003) by Margaret Atwood; Deadstock (2007) by Jeffrey Thomas; Accelerando (2005) by Charles Stross; Ware Tetralogy by Rudy Rucker; and Divergent (2011) by Veronica Roth.

In film, artificial meat has featured prominently in Giulio Questi's 1968 drama La morte ha fatto l'uovo (Death Laid an Egg) and Claude Zidi's 1976 comedy L'aile ou la cuisse (The Wing or the Thigh). "Man-made" chickens also appear in David Lynch's 1977 surrealist horror, Eraserhead. Most recently, it was also featured prominently as the central theme of the movie Antiviral (2012).

The Starship Enterprise from the TV and movie franchise Star Trek apparently provides a synthetic meat or cultured meat as a food source for the crew,[108] although crews from The Next Generation and later use replicators.

In the ABC sitcom Better Off Ted (20092010), the episode "Heroes" features Phil (Jonathan Slavin) and Lem (Malcolm Barrett) trying to grow cowless beef.

In the videogame Project Eden, the player characters investigate a cultured meat company called Real Meat.

In the movie "GalaxyQuest", during the dinner scene, Tim Allen's character refers to his steak tasting like "real Iowa beef".

Cultured meat was a subject on an episode of the Colbert Report on 17 March 2009.[109]

In February, 2014, a biotech startup called BiteLabs ran a campaign to generate popular support for artisanal salami made with meat cultured from celebrity tissue samples.[110] The campaign became viral on Twitter, where users tweeted at celebrities asking them to donate muscle cells to the project.[111] Media reactions to BiteLabs variously identified the startup as a satire on startup culture,[112] celebrity culture,[113] or as a discussion prompt on bioethical concerns.[114] While BiteLabs claimed to be inspired by the success of Sergey Brin's burger, the company is seen as an example of critical design rather than an actual business venture.

In late 2016, cultured meat was involved in a case in the episode "How The Sausage Is Made" of CBS show Elementary.

Originally posted here:
Cultured meat - Wikipedia

Posted in Iowa Stem Cells | Comments Off on Cultured meat – Wikipedia

Stem Cell – Joint Preservation Institute of Iowa

Posted: August 9, 2018 at 5:41 am

Orthobiologic is the broad term for using biologic products in orthopaedic surgeryranging from stem cell injections to matched donor bone and cartilage transplantation. This is an exciting and promising new field in orthopaedic surgery and Dr. Richard Goding offers all of the newest, highest technology treatments, as well as the time-tested gold standards.

Each patient and each joint has its own story, and what may be the best treatment for one patient may not be the most appropriate for another patient.Dr. Richard Goding is one of the few orthopaedic surgeons in the nation who offers the full spectrum of treatments in this field. He offers each patient a personalized plan that goes beyond the initial treatment Dr. Richard Goding remains involved in your care until your joint problem is solved.

The Joint Preservation Institute of Iowa is now offering stem cell therapy in Des Moines.

Dr. Richard Goding is one of the firstorthopaedic surgeons in the nation to offer this treatment. He uses autologous stem cell therapy and the regenerative power of mesenchymal stem cells to give patients another treatment option for dealing with orthopaedic conditions, such as osteoarthritis and other degenerative joint conditions.Read More >

MACI is a new procedure that treats the articular cartilage defects of the knee by assisting regeneration of cartilage and restoring flexibility. Articular cartilage is a tissue that covers the surface of the joints and is responsible for pain-free movement of the bones within the joint. If the articular cartilage is damaged, the ends of the bones rub against each other, causing pain. MACI is indicated for patients with significant cartilage defects causing joint pain, swelling and catching in the knee.Read More >

Osteoarticular transfer system (OATS) is a surgical procedure used to treat isolated cartilage defects, which are usually 10 to 20 milimeters in size. The procedure transfers cartilage plugs taken from non-weight bearing areas of the joint and to the damaged areas of the joint.Read More >

DeNovo grafts are tissue grafts used in cartilage repair. These grafts consist of cartilage tissue collected from donors or grown in the laboratory using human donated cartilage cells. There are two forms DeNovo ET (engineered tissue) and DeNovo NT (natural tissue).Read More >

Chondrofix Osteochondral Allograft takes the repair of full-thickness osteochondral lesions to a new level of convenience. Chondrofix is the first off-the-shelf osteochondral allograft, and each graft combines the inherent qualities of donated human bone and cartilage with the advantages of simplicity and safety. It is intended for homologous use to repair osteochondral lesions in diarthrodial joints. Read More >

Subchondroplasty is a minimally invasive, fluoroscopically assisted procedure that targets and fills subchondral bone defects through the delivery of AccuFill BSM, a highly porous, nanocrystalline injectable calcium phosphate.Read More >

Superior capsular reconstruction is a new procedure for treating large unfixable rotator cuff tears. This procedure, which is done on an outpatient basis almost fully arthroscopically, uses a graft to reconstruct the shoulder capsule when the rotator cuff tendon tears are too large to repair. By reconstructing the capsule, a cushion is placed between the ball of the shoulder joint and the acromion bone. Additionally, the joint is held in anatomic position, allowing for restoration of normal shoulder function.Read More >

Total shoulder replacement is a very successful procedure performed for shoulder arthritis. However, there are some significant limitations. The shoulder replacement does not tolerate heavy use and will wear out over time, making it a poor choice for younger patients. Read More >

Read more:
Stem Cell - Joint Preservation Institute of Iowa

Posted in Iowa Stem Cells | Comments Off on Stem Cell – Joint Preservation Institute of Iowa

Stem cell therapy and healing meniscus damage – Dr. Marc …

Posted: July 29, 2018 at 10:44 pm

Marc Darrow MD, JD. Thank you for reading my article. You can ask me your questions about stem cells and meniscus injury using the contact form below.

Often I will hear remarkable statements in emails. Here is an example of one shared about knee pain.

This person contacted me because they were interested in a stem cell program to help them avoid a knee replacement. He reported that he had an arthroscopic surgery to remove part of his meniscus recently. The doctor who performed the surgery told his patient to make sure he called him when they were ready for knee replacement.The doctor said that he was already familiar with this patients knee anatomy and he could be counted on to do a good job when it was time for the replacement.

As the predicted knee degeneration occurred, the patient instead reached out to us and stem cell therapy. Hindsight and foresight, the person had wished they would have reached out before the surgery so we could have helped with his meniscus repair.

New research into the healing world of the knee meniscus is fascinating. Despite decades of traditional medical beliefs that because of its poor or even absent network of blood vessels and blood supply, parts of the knee meniscus cannot heal. Researchers are discovering the meniscus is in fact, always trying to heal itself.

Can a meniscus really regenerate itself? This is a question I am often asked. The answer is yes, in some circumstance.

In 2011, three doctors from the North Middlesex University Hospital in the United Kingdom published a strange case history in the medical journal Case Reports in Medicine. What was odd about this case was that a 70 year old man who had recently undergone total knee replacement was suffering from terrible knee pain. It wasnt the knee pain that was odd, patients frequently report knee pain after knee replacement. What was strange to the doctors was what was causing the knee pain. Here is their case:

Report: The patients surgery had taken place at another hospital, and he had made an initially uneventful recovery with a good clinical range of motion and satisfactory postoperative radiographs. At 9 months, however, he began to develop medial and lateral retinacular (middle and side tendon related pain) and deep knee pain, without associated knee swelling, warmth, or wound disturbance. (No infection or apparent injury) His symptoms steadily worsened, particularly with load-bearing activity and bending his knee past 80 degrees.

Meniscus tissue was growing inside the knee replacement

Our patient was found to have soft-tissue entrapment between the femoral component and the polyethylene tray . . . suggestive of meniscus-like tissue. Meniscal regeneration has been previously described in experimental and clinical studies following meniscectomy and has also been previously reported following TKA (total knee replacement).(1)

Studies suggest that this is possible because the meniscus is always trying to heal itself. How did this patient story end? Another surgery, arthroscopic debridement to remove the new meniscus tissue because it was trapped in the hardware.

What was the moral of the story? Make sure you do not leave any meniscus behind in total knee replacement, it may regrow itself, and a70 year old man was not too old to grow new meniscus tissue and his own stem cells had something to do with it.

A new study in the Journal of orthopaedic research lead by the Department of Orthopaedics and Rehabilitation, University of Iowa discusses how a meniscus regenerates and heals.

The researchers hypothesized that the meniscus contains a population of regenerative cells, (cells that stimulate stem cell activity) and that they migrate to the site of meniscal injury. In the above study that is what the doctors speculated happened to their patient.

However, studies revealed that migrating cells were mainly confined to the red zone in normal menisci: (This is the area where the meniscus has good blood flow and healing elements are abundant). However, these cells were capable of repopulating defects made in the white zone, (the area without circulation). When the meniscus was injured, migrating cell numbers increased dramatically. Stem cells in the knee increased in number to combat the injury.These findings demonstrate that, much as in articular cartilage, injuries to the meniscus mobilize an intrinsic progenitor cell population with strong reparative potential, even into the white zone area.(2)

The short of it? The meniscus figures out how to heal itself if it can. Even in the areas that are typically believed unhealable because of lack of blood flow to that area.

Stem cell numbers?What is even more fascinating is that the meniscus signals for more stem cells from the knee capsule to come to the injured area. For those people asking about stem cell numbers that are harvested for treatment, you wont get an answer from the meniscus because it is mobilizing the stem cells already in the knee. Sometimes the meniscus only needs little spark, an injection of own stem cells to facilitate an abundant healing.

If you had a meniscus tear you are familiar with White Zone, and Red Zone, meniscus tears.The Red Zone, part of the meniscus, the outer edges, receives a steady stream of healing cells from its well organized blood vessel network. For those of you with a meniscus injury that is being recommended to surgery, you may have had your doctor explain to you that you have a White Zone, tear. The White Zone, lies in the center of the meniscus. It does not have a well organized blood network. It is these meniscal injuries that send patients to surgery. Unfortunately because of the lack of blood supply the damaged tissue needs to be removed.

In the Journal of orthopaedic research doctors examined the process of meniscal regeneration and cartilage degeneration following meniscus surgical removal in mice. They found that there is ahealing environment that the meniscus and cartilage create independently of each other spurred on by native stem cells, that later melds together, suggestive of a balance between meniscal regeneration and cartilage homeostasis.(3)The meniscus and cartilage are trying to regenerate each other.

This special relationship between cartilage, meniscus and stem cells is discussed in new research from the University of Iowa. The Iowa findings demonstrate that, much as in articular cartilage, injuries to the meniscus mobilize an intrinsic progenitor (stem cell) population with strong reparative potential.(4)The problem for patients is that despite the desire to heal and regenerate, as pointed out by the Iowa researchers, Serious meniscus injuries seldom heal and increase the risk for knee osteoarthritis; thus, there is a need to develop new reparative therapies. In that regard, stimulating tissue regeneration by autologous stem/progenitor cells has emerged as a promising new strategy.(4)

In past articles I have written extensively about how stem cells change the environment of diseased joints to healing. Research like that above confirms that when one part of the knee is repairing, the entire knee is repairing. This change of environment is something a surgery cannot offer.

Doctors at the University of Southern Denmarkconfirm this:

Patients undergoing arthroscopic partial meniscectomy are at increased risk of knee osteoarthritis. Meniscal damage and/or Meniscal surgery may alter knee-joint loading (the distribution of weight across the knee) to increase osteoarthritis risk.(5)

As noted, research has suggested poor long-term outcomes for patients with meniscectomies with increased incidence of osteoarthritis, leaving a need to develop technology to regenerate meniscal tissue following meniscectomy. The answer is stem cells.(6)

As you have seen in this article, the meniscus has a remarkable ability to heal itself. When someone comes into our office with knee problems we start with a conversation so we can learn about the patients lifestyle and what are his/her goals of the treatment. Is it to get back to marathon training or is it to get up and down a staircase without his/her knee locking up? Then we will do a detailed physical examination looking for those signs that will tell us how helpful stem cell therapy may be.

STEM CELL INSTITUTEA leading provider of bone marrow derived stem cell therapy, Platelet Rich Plasma and Prolotherapy in Los Angeles and the world!11645 WILSHIRE BOULEVARD SUITE 120, LOS ANGELES, CA 90025PHONE: (800) 300-9300

1 Matar HE, Dala-Ali B, Atkinson HD. Meniscal regeneration: a cause of persisting pain following total knee arthroplasty. Case reports in medicine. 2011;2011.

2 Seol D, Zhou C, Brouillette MJ, Song I, Yu Y, Choe HH, Lehman AD, Jang KW, Fredericks DC, Laughlin BJ, Martin JA. Characteristics of meniscus progenitor cells migrated from injured meniscus. Journal of Orthopaedic Research. 2016 Nov 1.

3 Hiyama K, Muneta T, Koga H, Sekiya I, Tsuji K.Meniscal regeneration after resection of the anterior half of the medial meniscus in mice.J Orthop Res. 2016 Nov 2. doi: 10.1002/jor.23470. [Epub ahead of print]

4 Seol D et al. Characteristics of meniscus progenitor cells migrated from injured meniscus. J Orthop Res. 2016 Nov 3. doi: 10.1002/jor.23472.

5Thorlund JB, Holsgaard-Larsen A, Creaby MW, et al.Changes in knee joint load indices from before to 12 months after arthroscopic partial meniscectomy: a prospective cohort study. Osteoarthritis Cartilage. 2016 Jul;24(7):1153-9. doi: 10.1016/j.joca.2016.01.987. Epub 2016 Feb 2.

6. McCrum CL, Vangsness CT. Postmeniscectomy Meniscus Growth With Stem Cells: Where Are We Now? Sports Med Arthrosc. 2015 Sep;23(3):139-42. doi: 10.1097/JSA.0000000000000073.

Original post:
Stem cell therapy and healing meniscus damage - Dr. Marc ...

Posted in Iowa Stem Cells | Comments Off on Stem cell therapy and healing meniscus damage – Dr. Marc …

The importance of stem cell numbers in stem cell therapy …

Posted: July 17, 2018 at 12:48 pm

Are there enough stem cells in your knees to heal the damage of osteoarthritis? If yes, why arent those stem cells fixing your knees now? Is it a lack of numbers? Or is it a lack of communication?

Marc Darrow MD, JD. Thank you for reading my article. You can ask me your questions about bone marrow derived stem cells using the contact form below.

In 2011, doctors at the University of Aberdeen published research in the journal Arthritis and rheumatism that provided the first evidence that resident stem cells in the knee joint synovium underwent proliferation and chondrogenic differentiation following injury.

In other words the number of stem cells in an injured knee increased and began to turn themselves into cartilage.(1)This paper, presenting the idea that stem cells in an injured knee increased in numbers in preparation of healing has been cited by more than 40 medical studies.

If the stem cells in the knee synovial lining are abundant and have the ability to rebuild cartilage after injury, why isnt the knee fixing itself?

One of those 40 studies was performed by researchers at theUniversity of Calgary in 2012. Among their questions, if the stem cells in the knee synovial lining are abundant and have the ability to rebuild cartilage after injury, why isnt the knee fixing itself? Here is what they published:

Since osteoarthritis leads to a progressive loss of cartilage and synovial progenitors (rebuilding) cells have the potential to contribute to articular cartilage repair, the inability of osteoarthritis synovial fluid Mesenchymal progenitor cells (stem cell growth factors) to spontaneously differentiate into chondrocytes suggests that cell-to-cell aggregation and/or communication may be impaired in osteoarthritis and somehow dampen the normal mechanism of chondrocyte replenishment from the synovium or synovial fluid. Should the cells of the synovium or synovial fluid be a reservoir of stem cells for normal articular cartilage maintenance and repair, these endogenous sources of chondro-biased cells would be a fundamental and new strategy for treating osteoarthritis and cartilage injury if this loss of aggregation & differentiation phenotype can be overcome.(2)

This research was supported in anew study from December 2017 In Nature reviews. Rheumatology. The paper suggested that recognizing that joint-resident stem cells are comparatively abundant in the joint and occupy multiple niches (from the center of the joint to the out edges) will enable the optimization of single-stage therapeutic interventions for osteoarthritis.(3) The idea is to get these native stem cells to repair.

Now we know that there are many stem cells in the knee, when there is an injury there are more stem cells. If we can figure out how to get these stem cells turned on to the healing mode, the knee could heal itself of early stage osteoarthritis. So the problem is not the number of stem cells, BUT, communication.

This failure to communicate was also seen in other research. In 2016, another heavily cited paper, this time fromTehran University for Medical Sciences, noted that despite their larger numbers,the native stem cells act chaotically and are unable to regroup themselves into a healing mechanism and repair the bone, cartilage and other tissue. Introducing bone marrow stem cells into this environmentgets the native stem cells in line and redirects them to perform healing functions. The joint environmentis changed from chaotic to healing because of communication.(4)

A recentpaper from a research team inAustralia confirms how this change of joint environment works. It starts with cell signalling a new communication network is built.

University of Iowa research published in theJournal of orthopaedic research

Serious meniscus injuries seldom heal and increase the risk for knee osteoarthritis; thus, there is a need to develop new reparative therapies. In that regard, stimulating tissue regeneration by autologous (from you, not donated) stem/progenitor cells has emerged as a promising new strategy.

(The research team) showed previously that migratory chondrogenic progenitor cells (mobile cartilage growth factors) were recruited to injured cartilage, where they showed a capability in situ (on the spot) tissue repair. Here, we tested the hypothesis that the meniscus contains a similar population of regenerative cells.

Explant studies revealed that migrating cells were mainly confined to the red zone (where the blood is and its growth factors) in normal menisci: However, these cells were capable of repopulating defects made in the white zone (the desert area where no blood flows. Migrating cell numbers increased dramatically in damaged meniscus. Relative to non-migrating meniscus cells, migrating cells were more clonogenic, overexpressed progenitor cell markers, and included a larger side population. (They were ready to heal) Gene expression profiling showed that the migrating population was more similar tochondrogenic progenitor cells (mobile cartilage growth factors) than other meniscus cells. Finally, migrating cells equaledchondrogenic progenitor cells in chondrogenic potential, indicating a capacity for repair of the cartilaginous white zone of the meniscus. These findings demonstrate that, much as in articular cartilage, injuries to the meniscus mobilize an intrinsic progenitor cell population with strong reparative potential.(6)

The intrinsic progenitor cell population with strong reparative potential are in your knee waiting to be mobilized.

A leading provider of bone marrow derived stem cell therapy, Platelet Rich Plasma and Prolotherapy11645 WILSHIRE BOULEVARD SUITE 120, LOS ANGELES, CA 90025

PHONE: (800) 300-9300

1 Kurth TB, Dellaccio F, Crouch V, Augello A, Sharpe PT, De Bari C. Functional mesenchymal stem cell niches in adult mouse knee joint synovium in vivo. Arthritis Rheum. 2011 May;63(5):1289-300. doi: 10.1002/art.30234.

2 Krawetz RJ, Wu YE, Martin L, Rattner JB, Matyas JR, Hart DA. Synovial Fluid Progenitors Expressing CD90+ from Normal but Not Osteoarthritic Joints Undergo Chondrogenic Differentiation without Micro-Mass Culture. Kerkis I, ed.PLoS ONE. 2012;7(8):e43616. doi:10.1371/journal.pone.0043616.

3 McGonagle D, Baboolal TG, Jones E. Native joint-resident mesenchymal stem cells for cartilage repair in osteoarthritis. Nature Reviews Rheumatology. 2017 Dec;13(12):719.

3Davatchi F, et al. Mesenchymal stem cell therapy for knee osteoarthritis: 5 years follow-up of three patients. Int J Rheum Dis. 2016 Mar;19(3):219-25. 2. Freitag J, Bates D, Boyd R, Shah K, Barnard A, Huguenin L, Tenen A.Mesenchymal stem cell therapy in the treatment of osteoarthritis: reparative pathways, safety and efficacy a review.BMC Musculoskelet Disord. 2016 May 26;17(1):230. doi: 10.1186/s12891-016-1085-9. Review.

4 Freitag J, Bates D, Boyd R, et al. Mesenchymal stem cell therapy in the treatment of osteoarthritis: reparative pathways, safety and efficacy a review.BMC Musculoskeletal Disorders. 2016;17:230. doi:10.1186/s12891-016-1085-9.

5 Seol D, Zhou C, et al. Characteristics of meniscus progenitor cells migrated from injured meniscus. J Orthop Res. 2016 Nov 3. doi: 10.1002/jor.23472.

Original post:
The importance of stem cell numbers in stem cell therapy ...

Posted in Iowa Stem Cells | Comments Off on The importance of stem cell numbers in stem cell therapy …

Blood and Marrow Transplant Program | University of Iowa …

Posted: July 15, 2018 at 1:45 am

The Blood and Marrow Transplant Program at University of Iowa Hospitals and Clinics in Iowa City, Iowa has been performing stem cell transplants since 1980. The program has earned reaccreditation from the Foundation for the Accreditation of Cellular Therapy (FACT), putting Iowa's BMT program on a select list of institutions that meet the most rigorous standards in every aspect of stem cell therapy, including clinical care, donor management, cell collection, processing, storage, transportation, administration, and cell release.

A bone marrow transplant is a procedure to replace damaged or destroyed bone marrow with healthy bone marrow stem cells. Bone marrow is the soft, fatty tissue inside your bones. Stem cells are immature cells in the bone marrow that give rise to all of your blood cells.

There are several alternative names for a blood and marrow transplant, including:

Our experienced team has provided over 3300 stem cell transplantsadult,pediatric, allogeneic and autologoussince 1980. Learn more about why the UI Blood and Marrow Transplant Program is your best choice for care.

There are three kinds of bone marrow transplants:

The term auto means self. Stem cells are removed from you before you receive high-dose chemotherapy or radiation treatment. The stem cells are stored in a freezer (cryopreservation). After high-dose chemotherapy or radiation treatments, your stems cells are put back in your body to make (regenerate) normal blood cells. This is called a rescue transplant.

The term allo means other. Stem cells are removed from another person, called a donor. Most times, the donor's genes must at least partly match your genes. Special blood tests are done to see if a donor is a good match for you. A brother or sister is most likely to be a good match. Sometimes parents, children, and other relatives are good matches. Donors who are not related to you may be found through national bone marrow registries.

This is a type of allogeneic transplant. Stem cells are removed from a newborn baby's umbilical cord right after birth. The stem cells are frozen and stored until they are needed for a transplant. Umbilical cord blood cells are very immature so there is less of a need for matching. But blood counts take longer to recover.

Before the transplant, chemotherapy, radiation, or both may be given. This may be done in two ways:

High-dose chemotherapy, radiation, or both are given to kill any cancer cells. This also kills all healthy bone marrow that remains, and allows new stem cells to grow in the bone marrow.

Patients receive lower doses of chemotherapy and radiation before a transplant. This allows older patients, and those with other health problems to have a transplant.

A stem cell transplant is done after chemotherapy and radiation is complete. The stem cells are delivered into your bloodstream usually through a tube called a central venous catheter. The process is similar to getting a blood transfusion. The stem cells travel through the blood into the bone marrow. Most times, no surgery is needed.

Adult solid tumors

Pediatric solid tumors

Most patients receiving a stem cell transplant are in their fifties and sixties. Sometimes, they are a little older. Transplantation is a very intensive therapy, and we need to have some reassurance that patients have a good chance of surviving and will not succumb to such intensive therapy. We try to maximize success and minimize the risk of patients actually having their life shortened by a stem cell transplant.

To assess the fitness of a patient to receive a transplant, we test:

In addition, we will ensure that there is no active infection by doing blood work and imaging tests such as CT chest, CT sinuses and/or PET-CT scan. These tests are arranged prior to the first clinic visit so that the results can be reviewed and decisions can be made about treatment options during the visit with the specialist.

The transplant process is outlined in detail in our Allogeneic and Autologous Guidebooks and instructional videos.

Original post:
Blood and Marrow Transplant Program | University of Iowa ...

Posted in Iowa Stem Cells | Comments Off on Blood and Marrow Transplant Program | University of Iowa …

Regenexx Des Moines | Iowa’s Regenexx Provider

Posted: October 15, 2017 at 9:16 am

Select a Problem Area

If you have pain, we're here to help. Regenexx Procedures are patented stem cell and blood platelet procedures that are used to treat a wide range of joint and spine conditions.

Click a problem area to discover what Regenexx can do for you.

The Regenexx family of non-surgical stem-cell & blood platelet procedures are next generation regenerative injection treatments for those who are suffering from shoulder pain due to arthritis, rotator cuff and shoulder labrum tears, overuse injuries, and other degenerative conditions. Regenexx is also a viable alternative for those considering shoulder replacement surgery.

View Details About Shoulder Treatments

Commonly Treated Conditions:

Shoulder Procedure Video

Regenexx Procedures are advanced stem cell and blood platelet procedures for foot and ankle conditions. Before you consider ankle surgery, fusion or replacement, consider the worlds leading stem cell and prp injection treatments.

View Details About Foot & Ankle Treatments

Commonly Treated Conditions:

Ankle Procedure Video

The Regenexx family of non-surgical stem-cell & blood platelet procedures are next generation regenerative injection treatments for those who are suffering from pain or reduced range of motion due to basal joint / cmc arthritis, hand arthritis, or other injuries & conditions in the hand.

View Details About Hand & Wrist Treatments

Commonly Treated Conditions:

The Regenexx family of non-surgical stem cell and blood platelet procedures offer next-generation injection treatments for those who are suffering from knee pain or may be facing knee surgery or knee replacement due to common injuries, arthritis, overuse and other conditions.

View Details About Knee Treatments

Commonly Treated Conditions:

ACL Procedure VideoIn-Depth with Dr. John Schultz ACL Procedure Video

The Regenexx family of non-surgical stem-cell & blood platelet procedures are next generation regenerative injection treatments for those who are suffering from pain, inflammation or reduced range of motion due tocommon elbow injuries, arthritis and overuse conditions.

View Details About Elbow Treatments

Commonly Treated Conditions:

The Regenexx family of hip surgery alternatives are breakthrough, non-surgical stem-cell treatments for people suffering from hip pain due to common injuries, hip arthritis & other degenerative problems related to the hip joint.

View Details About Hip Treatments

Commonly Treated Conditions:

Hip Labrum Procedure Video Hip Avascular Necrosis Procedure Video

Regenexx has many non-surgical platelet and stem cell based procedures developed to help patients avoid spine surgery and high dose epidural steroid side effects. These procedures utilize the patients own natural growth factors or stem cells to treat bulging or herniated discs, degenerative conditions in the spine, and other back and neck conditions that cause pain.

View Details About Spine Treatments

Commonly Treated Conditions:

Intradiscal Procedure Video

Regenexx has many non-surgical platelet and stem cell based procedures developed to help patients avoid spine surgery and high dose epidural steroid side effects. These procedures utilize the patients own natural growth factors or stem cells to treat bulging or herniated discs, degenerative conditions in the spine, and other back and neck conditions that cause pain.

View Details About Spine Treatments

Commonly Treated Conditions:

Cervical Spine Video

See original here:
Regenexx Des Moines | Iowa's Regenexx Provider

Posted in Iowa Stem Cells | Comments Off on Regenexx Des Moines | Iowa’s Regenexx Provider

Page 21234..»