Author Archives: admin


Weekly pick of Neuroscience news from around the world – Brain Tumour Research

The big news story for the cancer community this week has been the setting up of a new transatlantic research alliance with the ambition to develop new strategies and technologies to detect cancer at its earliest stage.

Those involved in this initiative, including Cancer Research UK (CRUK) believe thatearly detection is essential to help more people beat cancer a patients chance of surviving their disease improves dramatically when cancer is found and treated earlier.

Early diagnosis is, of course, a wish of the brain tumour community. All too often we hear of patients who have had to wait many months, with many visits to the GP before establishing the cause of their symptoms. However, we must not forget that earlier diagnosis may bring relief but there remains a lack of treatments. There is still no cure for many brain tumour patients

In the Report of the Task and Finish Working Group on Brain Tumour Research released February 2018 this was summed up perfectly by brain tumour activist Peter Realf who said While I endorse the need to improve earlier diagnosis, this alone without a cure will simply mean that patients face a longer walk to the grave.

In Texas there is work on in vitro blood brain barrier (BBB) models to equate their strengths and weaknesses. In-vitro means in the glass so these models are constructed with microorganisms, cells, or biological molecules outside their normal biological context e.g. in the petri dish or test tube. Work in this arena has previously been under taken at our University of Portsmouth centre. A combinatorial approach of in vitro BBB models and in-vivo (within the living) methods is thought to be key to the development of CNS therapeutics (medicines) with improved pharmacokinetic (the movement refers to the movement of drug into, though, and out of the body) properties and better BBB penetrability.

Most cancers kill because tumour cells spread, or metastasise, beyond the primary site, for example breast, to invade other organs, brain being one. Now, a University of Southern California (USC), study has found that circulating tumour cells in the blood target a particular organ and this knowledge may enable the development of treatments to prevent the spread of these metastatic cancers.

Analysis of these cells identified regulator genes and proteins within the cells that apparently directed the cancers spread to the brain. The team were therefore able to predict that a patients breast cancer cells would eventually migrate to the brain.

Assistant professor of stem cell and regenerative medicine at the Keck School of Medicine at USC, Min Yu, also discovered that a protein on the surface of these brain-targeting tumour cells helps them to breech the blood brain barrier and lodge in brain tissue, while another protein inside the cells shield them from the brains immune response, enabling them to grow there.

We can imagine someday using the information carried by circulating tumour cells to improve the detection, monitoring and treatment of the spreading cancers, Yu said.

A compound effective in killing chemotherapy-resistant glioblastoma-initiating cells (GICs) has been identified, raising hopes of producing drugs capable of eradicating refractory tumours (tumours that dont respond to treatment) with low toxicity.

As we are all too aware, despite longstanding and earnest endeavours to develop new remedies, the prognosis of most glioblastoma patients undergoing chemotherapies and radiotherapies remains poor with a median survival period of approximately 15 months.

One of the reasons for this is the lack of methods to eradicate its cancer stem cells, or glioblastoma-initiating cells (GICs), that demonstrate tumourigenicity (ability to form tumours) and resistance to chemotherapies and radiotherapies.

This study successfully cultured human GICs resistant to temozolomide (TMZ), the gold standard chemotherapy drug used for treating glioblastoma.

Then a high-throughput drug screening was conducted to identify a compound that could specifically kill or inflict damage to GICs, but not normal cells such as neural stem cells and astrocytes.

Compound 10580 was successfully identified as being capable of killing or inflicting damages to GICs whilst at the same time exhibiting no visible toxicity

"Compound 10580 is a promising candidate for developing drugs against glioblastoma and other recurring cancerssaid Toru Kondo of Hokkaido University's Institute for Genetic Medicine who led the study.

What is also interesting here is the collaborative nature of the study group with Hokkaido University, working alongside FUJIFILM Corporation and the National Institute of Advanced Industrial Science and Technology (AIST).

Related reading:

If you found this story interesting or helpful,sign up to our weekly e-newsand keep up to date with all the latest from Brain Tumour Research.

Excerpt from:
Weekly pick of Neuroscience news from around the world - Brain Tumour Research

Cesca Therapeutics Forms Joint Venture with Healthbanks Biotech (USA) to Provide Immune Cell Banking and Cell Processing Services – PRNewswire

RANCHO CORDOVA, Calif., Oct. 22, 2019 /PRNewswire/ -- Cesca Therapeutics Inc.(Nasdaq: KOOL), a market leader in automated cell processing and autologous cell therapies for regenerative medicine, and ThermoGenesis, its wholly owned device subsidiary, today announced that the company has entered into a definitive joint venture agreement with HealthBanks Biotech (USA) Inc., one of the world's leading stem cell bank networks, to commercialize its proprietary cell processing platform, CAR-TXpress, for use in immune cell banking as well as for cell-basedcontract development and manufacturing services (CMO/CDMO). The joint venture will be named ImmuneCyte Life Sciences Inc. ("ImmuneCyte") and is expected to officially launch during the fourth quarter of 2019.

Under terms of the agreement, ImmuneCyte will initially be owned 80% by HealthBanks Biotech and 20% by Cesca. Cesca will contribute to ImmuneCyte exclusive rights to use ThermoGenesis' proprietary cell processing technology for the immune cell banking business and non-exclusive rights for other cell-based contract development and manufacturing services. Cesca will also contribute its clinical development assets to the joint venture, as the company has decided to discontinue these activities in order to focus exclusively on the device business.

Once operational, ImmuneCyte will be among the first immune cell banks in the U.S. to provide clients with the opportunity to bank their own healthy immune cells for future use as a resource for cell-based immunotherapies, such as dendritic cell and chimeric antigen receptor (CAR) T-cell therapies. ImmuneCyte will utilize ThermoGenesis' proprietary CAR-TXpress platform which allows for the isolation of different components from 200 ml of blood in cGMP compliant, closed system. Given that the CAR-TXpress platform can increase cell processing efficiency by up to 16-fold as compared with the traditional, labor-intensive ficoll gradient centrifugation-based cell processing method, ImmuneCyte is expected to offer customers an unparalleled competitive advantage, including an ability to store their own immune cells at a tangibly lower cost.

"The ImmuneCyte joint venture will be paramount to the execution of our strategy to become a preferred cell processing and manufacturing solution provider in the cell and gene therapy field," said Dr. Chris Xu, Chairman and Chief Executive Officer of Cesca Therapeutics. "CAR-T therapeutic research is advancing rapidly. Partnering with HealthBanks Biotech, one of the foremost stem cell bank networks, with an experienced team and an established global infrastructure, will offer customers the ability to preserve younger, healthier and uncontaminated immune cells for potential future use. By applying our proprietary CAR-TXpress technology to immune cell banking and other CDMO cellular manufacturing services, we will allow for the manufacture and production of more effective and less costly immunotherapies."

In 2017, the U.S. Food and Drug Administration (FDA) approved two CAR-T cell therapies, under breakthrough designation, for the treatment of advanced B cell leukemia and lymphomas. Both use autologous (a patient's own) immune T cells to fight cancer and have reported an over 80% response rate in the "no-option" patient group, for those who have failed both chemo- and radiation therapies. This has helped to spur massive global interest for the development of additional CAR-T immunotherapies1. By the end of September 2019, there were over 800 CAR-T cell clinical trials registered on the http://www.clinicaltrials.gov website, targeting a wide variety of blood cancers and solid tumors.

Although highly effective, several recent studies on the eligibility of patients to enroll in CAR-T clinical trials showed that as many as 30-50% of cancer patients may not be eligible to enroll or to get sufficient CAR-T cells manufactured for the therapy. Reasons may include: (1) the function of the immune system declines with age and can be negatively affected by other medical conditions, (2) most standard cancer therapies, such as chemotherapy and radiation, destroy the immune system, and (3) in many cases of advanced cancer, cancer cells will enter circulation, invade and interfere with the body's natural production of immune cells. According to a recently reported JULIE trial, a CAR-T clinical trial in relapsed or refractory diffuse large B-cell lymphoma (DLBCL), one-third of the 238 screened patients failed to be enrolled, and more than half of the 238 failed to receive the intended CAR-T therapy2,3. ImmuneCyte will offer customers the ability to preserve younger, healthier and uncontaminated immune cells, for potential future use in advanced cancer immunotherapy.

About HealthBanks Biotech (USA) Inc.HealthBanks Biotech, headquartered in Irvine, CA, is one of the leading stem cell bank networks in the world and offers services globally through its sister companies located in the United States and other regions and nations. HealthBanks Biotech is accredited by the FDA, AABB, and CAP. The HealthBanks Biotech group was originally founded in 2001 with a vision that stem cells and cell and gene therapies could transform modern medicine. HealthBanks Biotech is a subsidiary of Boyalife Group, Inc. (USA), an affiliate of Boyalife (Hong Kong) Limited, the largest stockholder of Cesca. For more information about HealthBanks Biotech (USA) Inc., pleasevisit:www.healthbanks.us.

About ImmuneCyte Life Sciences Inc.ImmuneCyte will provide clients with the opportunity to bank their own immune cells when the cells are "healthy and unaffected" as a future resource for cellular immunotherapies, such as CAR-T. ImmuneCyte utilizes a proprietary CAR-TXpress platform, a GMP compliant close-system capable of automated separating and cryopreserving different components from blood.For more information about ImmuneCyte Life Sciences Inc., pleasevisit:www.immunecyte.com.

About Cesca Therapeutics Inc.Cesca Therapeuticsdevelops, commercializes and markets a range of automated technologies for CAR-T and other cell-based therapies. Its device division, ThermoGenesis develops, commercializes and markets a full suite of solutions for automated clinical biobanking, point-of-care applications, and automation for immuno-oncology. The Company has developed a semi- automated, functionally closed CAR-TXpressplatform to streamline the manufacturing process for the emerging CAR-T immunotherapy market. For more information about Cesca and ThermoGenesis, pleasevisit: http://www.cescatherapeutics.com.

Company Contact:Wendy Samford916-858-5191ir@thermogenesis.com

Investor Contact:Paula Schwartz,Rx Communications917-322-2216pschwartz@rxir.com

References:

1. Facts About Chimeric Antigen Receptor (CAR) T-Cell Therapy, Leukemia and Lymphoma Society (2018). https://www.lls.org

2. Updated Analysis of JULIET Trial: Tisagenlecleucel in Relapsed or Refractory DLBCL (2018).

3. Eligibility Criteria for CAR-T Trials and Survival Rates in Chemorefractory DLBCL. Journal of Clinical Pathways (2018).

SOURCE Cesca Therapeutics Inc.

http://www.cescatherapeutics.com

Read the original here:
Cesca Therapeutics Forms Joint Venture with Healthbanks Biotech (USA) to Provide Immune Cell Banking and Cell Processing Services - PRNewswire

Advancing patient care through innovative orthopaedics – SciTech Europa

Founded in 1958, the AO foundation is a medically guided, not-for-profit organisation led by an international group of surgeons specialised in the treatment of trauma and disorders of the musculoskeletal system. Today, the AO has a global network of over 200,000 health care professionals. Each year it offers over 830 educational events around the world, which are supported by nearly 9,000 faculty and are attended by over 58,000 participants. It has 20,000 surgeon members working in the fields of trauma, spine, craniomaxillofacial (CMF), veterinary, and reconstructive surgery.

The Mission of the AO foundation is promoting excellence in patient care and outcomes in trauma and musculoskeletal disorders. The focus of the AO clinical divisions, clinical unit, and Institutes, is on producing new concepts for improved fracture care, delivering evidence-based decision making, guaranteeing rigorous concept and product approval as well as timely and comprehensive dissemination of knowledge and expertise. The AO is made up from four clinical divisions (AOTrauma, AOSpine, AOCMF, AOVET), one clinical unit (AORecon), and four institutes the AO Research Institute Davos (ARI), AO Education Institute, AO Clinical Investigation & Documentation and AO Technical Commission (AOTK).

AO Research Institute Davos (ARI) is both the academic arm and the translational arm of the AO foundation. In its work to further the AO foundations mission (promoting excellence in patient care and outcomes in trauma and musculoskeletal disorders), ARIs purpose is to advance patient care through innovative orthopaedic research and development.

The goals of ARI include: Contribute high quality applied preclinical research and development (exploratory and translational) focused towards clinical applications/solutions; investigate and improve the performance of surgical procedures, devices and substances; foster a close relationship with the AO medical community, academic societies, and universities; and provide research environment / research mentorship / research support for AO clinicians.

Our Bone Regeneration focus area looks at bone healing in response to fracture involving a complex sequence of dynamic events, directed by numerous different cell types and growth factors. A critical factor for bone repair is the maintenance, or effective restoration, of an adequate blood supply, which is necessary to provide the damaged tissue with oxygen, nutrients and growth factors, as well as immune cells and mesenchymal stem cells required to repair the damage and induce new bone formation. Although bone generally has a high regenerative capacity, in some cases this inherent bone healing is compromised, which results in delaying healing or non-union of the bone fracture with increased health care costs and reduced quality of life issues for affected patients.

While a variety of risk factors have been identified that predispose a patient to an increased risk of developing delayed bone healing or non-union, it is currently not possible to identify specific at-risk patients at an early stage. Using in vitro, in vivo and microfluidic technologies, the aim of the Bone Regeneration Focus Area is to gain a greater understanding of the cellular interactions and mediators, including immunoregulation, underlying such impaired healing responses. By determining how cells such as immune cells, mesenchymal stem cells and endothelial cells normally interact during the repair process, and how this process is altered during impaired healing, we can then identify key mediators of the healing process. Our goal is to use tissue engineering and regenerative medicine approaches to promote bone healing, aimed at restoring bone integrity and its effective biomechanical properties.

In terms of this focus area, we aim at investigating the potential mechanisms leading to intervertebral disc (IVD) damage and evaluating novel biological treatment methods for IVD repair and regeneration. Acute and chronic damage to the IVD are major causes of low back pain. However, the factors that contribute to the loss of function of the IVD and the underlying pathophysiology are still poorly understood. We have established a whole IVD organ culture system with the ability to maintain entire discs with the endplates for several weeks under controlled nutrient and mechanical loading conditions.

Within this bioreactor, the beneficial or detrimental effects of nutrition, mechanical forces, and/or biochemical factors on disc cell viability and metabolic activity can be investigated. We have developed various defect and degeneration models, allowing us to design and evaluate appropriate biological treatment strategies. These include implantation of cells, delivery of anabolic, anti-catabolic or anti-inflammatory molecules, biomaterials or a combination thereof. Data from ex vivo models are also correlated to in vivo observations to identify molecular markers of IVD damage or degeneration.

To study the potential of new therapies for articular cartilage repair and regeneration, a bioreactor system applying multiaxial load to tissue-engineered constructs or osteo-chondral explants has been established. The bioreactor mimics the load and motion characteristics of an articulating joint. Chondral and osteochondral defect and disease models enable us to test tailored treatments under physiologically relevant mechanically loaded ex-vivo conditions. Cell- and material-based therapies as well as chondrogenic or anti-inflammatory factors are under investigation for cartilage repair and regeneration.

Biomaterials for skeletal repair can provide structural and mechanical features for the filling of defects, but also be a carrier for drugs, cells and biological factors. One of our goals is the development of 3D structures for bone, disc and cartilage tissue engineering, using tailored polymers and composites manufactured with additive manufacturing processes.

Our experience lies in the design of biocompatible, biodegradable polymers and their processing with controlled architecture and embedded biologics. A second field of research investigates the preparation of hyaluronan, a natural occurring biopolymer, based biomaterials which can be used to deliver drugs and cells. These injectable biodegradable materials have considerable potential in infection prophylaxis and tissues repair. We are also developing innovative technologies for the structuration and assembly of tissue-like matrices aiming to mimic for example, biological matrix mechanical and structural anisotropy. Additive manufacturing technologies will lead to the development of patient specific implants that can be tailor made to each individual case.

The Stem Cell focus area is particularly interested in stem cell therapies for bone and cartilage that could be applied within a clinical setting. We are increasingly investigating donor variation with the aim to predictively identify the potency of cells from individual donors. In the search for biomarkers to determine patient specific healing potential, exosomes and non-coding RNA sequences such as miRNA are increasingly being used as a diagnostic and therapeutic tool. The development of a serum-based biomarker approach would dramatically improve patient specific clinical decisions.

We also aim to investigate the role of mechanical and soluble factors in the activation of mesenchymal stem cells, and the promotion of differentiation and tissue repair. We can induce chondrogenic differentiation of human MSCs purely by mechanical stimulation and this is leading to new insights into cell behavior under loading conditions. Mechanical forces can be applied by way of rehabilitation protocols and are able to modify stem cell and immune cell function. Such studies are forming the basis of the emerging field of regenerative rehabilitation. In addition to the effect of load on direct differentiation, it is known that biomechanical stimulation can modulate the cell secretome. Investigating these changes could lead to the identification of new targets that may be present during articulation. This offers new avenues for potential clinical therapies.

The Musculoskeletal Infection team focusses their research activities on Fracture-Related Infection (FRI), with goals to optimise antibiotic prophylaxis, reduce the burden of therapeutic interventions, and study the impact of co-administered medication on infection. Our studies include preclinical in vitro and in vivo studies, as well as an increasing focus on observational studies in human patients.

In collaboration with ARI colleagues in the preclinical testing facility, we now have models that can mimic an open fracture, with a chronology and fixation that more accurately reflects clinical reality. Further advancements in our animal models in the past year include the controlled delivery of antimicrobials via the use of programmable, implantable pumps to more precisely control antibiotic dosing. In addition, we have investigated in more detail the use of anti-inflammatory medication in our animal studies and found it can have a major impact on treatment outcome, and so will be a focus for future studies with clear relevance for trauma patients. The preclinical evaluation of novel anti-infective interventions under Good Laboratory Practice (GLP) conditions has also continued in the past year, with two novel antimicrobial intervention studies performed in this space in the past year.

On the in vitro side, we have begun to develop an in vitro model for Staphylococcus aureus infection that has the potential to include human immune system cell-lines. This can not only reduce future animal studies but will also allow us to test interventions in a human-specific system. The antibiotic loaded hydrogel that has been in testing in ARI for several years, has now also been tested against MRSA biofilms and continues to be superior to aqueous solutions of antibiotics. In patient samples, we have made our first preparations for a study on the impact of antibiotic therapy on the human gut and skin microbiome. This is an under explored area of immense potential for bone health and will be a multi-year investigation with expert collaborations internationally.

A Fracture-Related Infection (FRI) consensus meeting in Davos in December 2016 achieved consensus on the fundamental features of FRI, and a proposal for defining the presence of FRI was reached. The establishment of this definition offers the opportunity to standardise preclinical research, improves the reporting of clinical studies and finally of course also aids in the decision-making during daily clinical practice. In the following 18 months, the expert group shifted attention to the next phase, validating the diagnostic criteria and develop treatment principles for FRI and a consensus on diagnosis and treatment principles for FRI.

In reflecting the greater complexity of this question, and to engage with other professional organisations, the group has grown to include external partners. Joining the ARI, AOTrauma and the AOTK Anti-Infection task force (AITF), is the European Bone and Joint Infection Society (EBJIS), the Orthopaedic Trauma Association (OTA), and the Pro-Implant Foundation, as well as a broadened panel of experts with extensive clinical experience in FRI. A first meeting of the expert group took place in Zrich in February. Prior to the meeting, the group was asked to review and consider the published literature on FRI, within nine specific concepts that were then presented for discussion in dedicated sessions during the meeting. The meeting engaged 35 experts and key opinion leaders in the field of FRI. Recommendations were developed on diagnosis and treatment of FRI. These guiding principles will be made available through scientific publications and an AO Bone Infection App.

Internal fracture fixation existed but only in individual hospitals and not globally, that is where ARI and AO came in and rolled this out globally and invented many new additions to this. ARI invented compression plates, minimal invasive surgery for trauma (plates, screws, nails etc.), locking plates for fractures close to articulating joints and for osteoporotic patients.

Currently tissue engineering and regenerative medicine (TERM) is in the research stage of its life cycle and has not really translated into routine surgical practice in orthopaedics. The combination of cells and biomaterials however has great potential in repair. The main issues are again regulatory, and the best way forward would be to develop techniques that can be applied in a single surgery within the operation room. Anything beyond this window and outside the operation room will take a significant amount of time to get approval and will likely not be surgeon friendly and obviously will be very costly.

TERM has its biggest potential in orthopaedics in the areas of cartilage repair (delaying classic orthopaedics), disc regeneration (back pain being one of the largest problems globally) and in bone this could be in large bone defects, but not a major area in fracture repair, where appropriate mechanical stimulation can be used to drive the repair to optimum levels and speed (which is also in the research stage). TERM has also potential in tendon and ligament repair.

Imaging and biomarkers for diagnostics and therapy (Theranostics) will be important in early detection of diseases or complications and then to prevent further development of the disease, delaying the time until classic orthopaedics is required. This may go beyond stopping the disease and towards tissue regeneration. The earlier the detection, the more potential for TERM.

The main challenges for a researcher are in translation and the fact that large companies today exist in a more complex regulatory environment, which means they are inclined to be very risk averse. This means in practice they need to see evidence of benefits or proof-of-concept in a clinical setting. The researchers in turn need to have greater awareness of these regulatory issues relating to medical development and CE approved manufacturers, than in the past. The increasingly complex regulatory environment of course has a greater impact on small companies and spin-offs, and can be seen as having a dampening effect on innovation development. Incremental innovations or solutions to niche problems will struggle to get the funding needed to carry them through the regulatory approval process. Researchers do benefit from this too, since in an environment in which companies are inclined to be more risk averse, they place a higher premium on solutions or concepts that have been through a rigorous clinical testing process. In orthopedics, we are approaching an innovation plateau with metals, and new technologies (such as tissue engineering, which is showing good results in research at present) still need to kick in to date little has translated to the patient in this field. 3D printing may have a place in spine or craniomaxillofacial areas, but offers little benefits to trauma in the most common areas for fracture repair. Surgeons who promote patient specific implants (PSI) in joint replacement have little proof that this offers clear improvements compared to current well-tested and proven joint replacement implants. The seamless integration of digitisation and robotic help into the patient treatment work-flow is another area to grow to help the surgeons in their daily practice.

Prof R. Geoff Richards

Director

AO Research Institute Davos

geoff.richards@aofoundation.org

Tweet @AOFoundation

https://www.aofoundation.org/Structure/research/exploratory-applied-research/research-institute/Pages/exploratory-applied-research.aspx

See the article here:
Advancing patient care through innovative orthopaedics - SciTech Europa

Worlds first cell atlas of developing liver created by Cambridge scientists – Cambridge Independent

The worlds first cell atlas of the human developmental liver has been created, giving fresh insight into how the blood and immune systems develop in the foetus.

A high-resolution resource, it will aid our understanding of normal development and efforts to tackle diseases that can form during development, such as leukaemia and immune disorders.

The cell atlas maps how the cellular landscape within the developing liver changes between the first and second trimesters of pregnancy, including how stem cell from the liver seed other tissues, supporting the high demand for oxygen required for growth.

Researchers from the Wellcome Sanger Institute in Hinxton, the Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Newcastle University and their collaborators created the atlas by using single cell technology to analyse 140,000 liver cells and 74,000 skin, kidney and yolk sac cells.

In adults, it is bone marrow that is primarily responsible for the creation of blood and immune cells in a process called haematopoiesis.

In early embryonic life, the yolk sac and liver play a key role in creating these cells, which then seed peripheral tissues such as skin, kidney and ultimately bone marrow.

But until now, the precise process of how blood and immune systems develop in humans has been unknown.

Isolating cells from the developing liver, the researchers were able to identify them by what genes they were expressing and discover what the cells looked like.

They tagged haematopoietic cells in sections of developmental liver using heavy metal markers in order to map them to their location.

Prof Muzlifah Haniffa, a senior author of the study from Newcastle University and senior clinical fellow at the Wellcome Sanger Institute, said: Until now research in this area has been a little bit like blindfolded people studying an elephant, with each describing just a small part of it.

This is the first time that anyone has described the whole picture, how the blood and immune systems develop in such detail. Its been an extraordinary, multidisciplinary effort that is now available as a tool for the whole scientific community.

The scientists learned that during foetal development, mother haematopoietic stem cells stay in the liver. But the liver alone cannot supply enough red blood cells, so the next generation daughter cells called progenitor cells travel to other tissues, maturing in places such as the skin. Thee, they develop into red blood cells to help meet the high demand for oxygen in the developing foetus.

Dr Elisa Laurenti, a senior author from the Wellcome MRC Cambridge Stem Cell Institute and the Department of Haematology at the University of Cambridge, said: We knew that as adults age our immune system changes. This study shows how the livers ability to make blood and immune cells changes in a very short space of time, even between seven and 17 weeks post-conception.

If we can understand what makes the stem cells in the liver so good at making red blood cells, it will have important implications for regenerative medicine.

The study, published in Nature, also involved the mapping of genes involved in immune deficiencies to reveal which cells were expressing them.

It is known that gene mutations can lead to immune disorders such as leukaemia.

A better understanding of the development of healthy liver functions could aid our understanding of how to treat such conditions.

The work is part of the ambitious effort to create the first complete Human Cell Atlas.

Dr Katrina Gold, genetics and molecular sciences portfolio manager at Wellcome, said: Our immune system is vital in helping to protect us from disease, yet we know very little about how immune cells develop and behave in the early embryo. This study is hugely important, laying a critical foundation for future research that could help improve our understanding of disorders linked to the early immune system, such as childhood leukaemias.

The Human Cell Atlas has the potential to transform our understanding of health and disease and were excited to see these first discoveries from our Wellcome-funded multidisciplinary team of scientists.

Dr Sarah Teichmann, a senior author from the Wellcome Sanger Institute, University of Cambridge and co-chair of the Human Cell Atlas organising committee, said: The first comprehensive cellular map of the developmental liver is another milestone for the Human

Cell Atlas initiative.

The data is now freely available for anyone to use and will be a great resource to better understand healthy cellular development and disease-causing genetic mutations.

Read more

Asthma treatment hope as Human Cell Atlas project creates first map of lungs

Sanger Institute scientist helps unveil blueprint for extraordinary Human Cell Atlas

AstraZeneca and Cancer Research UK launch joint Functional Genomics Centre in Cambridge

Go here to read the rest:
Worlds first cell atlas of developing liver created by Cambridge scientists - Cambridge Independent

Phenotypic Screening Advances in Technologies and Techniques – Technology Networks

Phenotypic screening is gaining new momentum in drug discovery with the hope that this approach will improve the success rate of drug approval.1 In this article we look at some of the latest screening tools and their applications.

This is illustrated by their recent study with Dr Ayman Zen where the team developed a high-content imaging screen using the endothelial tube formation assay, miniaturized to a 384-well plate format. Screening with an annotated chemical library of 1,280 bioactive small molecules identified a retinoid agonist, Tazarotene, that enhanced in vitro angiogenesis and wound healing in vivo. This high content screen identified an already FDA-approved small molecule that could be potentially exploited in regenerative medicine.3

Immuno-oncology: Pushing the Frontier of Discovery Through Advanced High Throughput Flow Cytometry

Immuno-oncology encompasses a number of approaches with one common thread: they harness the bodys own immune system against cancer.

Download this article to learn how advanced throughput flow cytometry overcomes these challenges to drive forward innovation in the immuno-oncology field.

Ebner is currently working in collaboration with recent Nobel-Prize winner Peter Ratcliffe, alongside scientists at Edinburgh University and MIT, to model hypoxia in glioblastoma. Hypoxia is a problem with some glioblastomas as it protects cells from radiotherapy treatment. Our aim is to use Peters expertise to help us set up an assay that mimics real tumor hypoxia. Then if we can identify small compounds that alter that hypoxic condition we can make the glioma cells more susceptible to either radiotherapy or temozolomide or some other treatment combination.

The labs main readout is high-content imaging, using fluorescent microscopy that can take many thousands of pictures. This approach utilizes different labels and harnesses software that automates the image analysis. The image analysis is set by the biologists but then it's applied across the entire screen. Its lower throughput than plate-based readout, but you get a lot more information out of the images, says Ebner. Increasingly, high content imaging is moving towards using AI and deep learning where you're trying to draw out even more information than the primary phenotype that you were looking at.

Indeed, a recent study using CRISPR-Cas9 mutagenesis showed that the proteins targeted by many cancer drugs currently in clinical development are non-essential for tumor growth, despite evidence to the contrary from previous studies using RNAi and small molecule inhibitors.4 In addition, the efficacy of the drugs tested was unaffected when CRISPR was used to knockout its assumed target suggesting that many are eliciting their anticancer activity through off-target effects.

The other benefit of CRISPR is that its extremely flexible, says Pettitt. This means you can expand the range of cell line models, for example, that you can screen in. The key reason why RNAi was such a popular technology, and now CRISPR is, is that you can basically knock out a gene by synthesizing just a short piece of RNA, he explains. CRISPR guides are very easy to synthesize, you can do it in a very high throughput setting, and you can design customized libraries to knock out every gene in the genome or a particular set of genes. As long as you can get the CRISPR machinery into your cells, it works very reliably.

The classic CRISPR (CRISPR-Cas9) system comprises a nuclease called Cas9 which you can program with a short RNA (20 nucleotides). The RNA will direct the nuclease to a certain site in the genome that matches and the nuclease will cleave the genome at that point. Repair of that double-strand break results in small insertions and deletions that result in knock out of a gene. But theres now more evolved applications of the technology emerging.

I think it's possible to be very creative with CRISPR in a way that it isnt with RNAi, says Pettitt. With RNAi you can really only shut genes off, but with CRISPR as well as making random mutations to knock out genes - you can also precisely edit genes if you provide a template region with a mutation with it. This can be incorporated into the target site for CRISPR so you can introduce the specific mutation youre interested in.

One such example is the problem with BRCA1 mutations: its important to be able to functionally classify whether these mutations are benign or pathogenic. A recent study used CRISPR to test 96.5% of all possible single-nucleotide variants (SNVs) in exons that encode functionally critical domains of BRCA1 and found over 400 non-functional missense SNVs were identified, as well as around 300 SNVs that disrupt expression. This knowledge will immediately aid clinical interpretation of BRCA1 genetic test results.5 In another study,6 Pettitt and colleagues used genome-wide CRISPR-Cas9 mutagenesis screens to identify the mutated forms of PARP that cause in vitro and in vivo PARP inhibitor resistance, and found that these mutations are also tolerated in cells with a pathogenic BRCA1 mutation resulting in a different profile of sensitivity to chemotherapy drugs compared with other types of PARP inhibitor resistance.

You couldnt screen at that level of detail using RNAi, where you design custom CRISPR that targets many different regions of the same gene and you can figure out which domains of the protein are important for your phenotype of interest, says Pettitt.

There are other evolutions of CRISPR now being developed as screens. For example, if you mutate the nuclease activity of Cas9, it still retains its ability to localize to the target site, so you can fuse Cas9 to transcriptional activators or repressors, and screen for transcriptional repression with CRISPR, as well as knock-out screens, says Pettitt. Theres also a whole range of CRISPR tools being developed that will edit bases by causing missense mutations rather than insertions or deletions, or causing methylation of DNA, or bringing in fluorescent proteins so you can visualize where the DNA sequences in the cells are. Its a measure of how flexible and useful CRISPR is in comparison to RNAi.

So will CRISPR be the one technology that everyone turns to for phenotypic screening in future? Im a firm believer that no technology answers every question, says Ebner. CRISPR is amazing, its use as a therapeutic or biologic is the stuff of science fiction. But as a tool for target identification, it comes with one important caveat. CRISPR knockout means exactly that it removes the potential protein that would otherwise be in the mix. Thats very different from a small compound inhibiting a protein that is still able to form a complex or that is just not active. Its the perfect example of a brilliant technology that is transformative, but it's not perfect. No technology is perfect.

References

1. Zheng W, Thorne N and McKew JC. Phenotypic screens as a renewed approach for drug discovery. Drug Discov. Today 2013; 18: 1067-1073.

2. Horvath P, Aulner N, Bickle M, et al. Screening out irrelevant cell-based models of disease. Nat Rev Drug Discov. 2016 Nov;15(11):751-769. doi: 10.1038/nrd.2016.175. Epub 2016 Sep 12.

3. Al Haj Zen A, Nawrot DA, Howarth A, et al. The Retinoid Agonist Tazarotene Promotes Angiogenesis and Wound Healing. Mol Ther. 2016 Oct;24(10):1745-1759. doi: 10.1038/mt.2016.153.

4.Lin et al. Off-target toxicity is a common mechanism of action of cancer drugs undergoing clinical trials. Science Translat Med. 2019; 11: (509). doi: 10.1126/scitranslmed.aaw8412

5.Findlay GM, Daza RM, Martin B et al. Accurate classification of BRCA1 variants with saturation genome editing. Nature. 2018 Oct; 562(7726): 217222. doi: 10.1038/s41586-018-0461-z

6.Pettitt et al. Genome-wide and high-density CRISPRCas9 screens identify point mutations in PARP1 causing PARP inhibitor resistance. Nat Commun. 2018 May 10;9(1):1849. doi: 10.1038/s41467-018-03917-2.

More here:
Phenotypic Screening Advances in Technologies and Techniques - Technology Networks

Can organoids, derived from stem cells, be used in disease treatments? – The Hindu

The story so far: On Monday, October 21, at Neuroscience 2019, the Society for Neurosciences 49th annual meeting, held in Chicago, U.S., two neuroscientists warned the gathering that fellow scientists are perilously close to crossing the ethical red line of growing mini-brains or organoids in the laboratory that can perceive or feel things. In some cases, scientists have already transplanted such lab-grown brain organoid to adult animals. The transplanted organoid had integrated with the animal brain, grown new neuronal connections and responded to light. Similarly, lung organoid transplanted into mice was able to form branching airways and early alveolar structures. These are seen as a step towards potential humanisation of host animals.

Organoids are a group of cells grown in laboratories into three-dimensional, miniature structures that mimic the cell arrangement of a fully-grown organ. They are tiny (typically the size of a pea) organ-like structures that do not achieve all the functional maturity of human organs but often resemble the early stages of a developing tissue. Most organoids contain only a subset of all the cells seen in a real organ, but lack blood vessels to make them fully functional. In the case of brain organoids, scientists have been able to develop neurons and even make specific brain regions such as the cerebral cortex that closely resemble the human brain. The largest brain organoids that have been grown in the laboratory are about 4 mm in diameter.

Organoids are grown in the lab using stem cells that can become any of the specialised cells seen in the human body, or stem cells taken from the organ or adults cells that have been induced to behave like stem cells, scientifically called induced pluripotent stem cells (iPSC). Stem cells are provided with nutrients and other specific molecules to grow and become cells resembling a specific organ. The growing cells are capable of self-organising into cellular structures of a specific organ and can partly replicate complex functions of mature organs physiological processes to regeneration and being in a diseased state.

Organoids of the brain, small intestine, kidney, heart, stomach, eyes, liver, pancreas, prostate, salivary glands, and inner ear to name a few have already been developed in the laboratory.

Since the use of embryonic stem cells to grow organs of interest has been mired in controversy leading to a ban on such research, researchers have turned to generating organoids using stem cells. Researchers have been successful in generating organoids of increasing complexity and diversity. Since the organoids closely resemble mature tissues, it opens up new vistas. These include studying the complex arrangements of cells in three-dimension and their function in detail, and understanding how cells assemble into organs.

Organoids can be used to study the safety and efficacy of new drugs and also test the response of tissues to existing medicines. Organoids will bring precision medicine closer to reality by developing patient-specific treatment strategies by studying which drugs the patient is most sensitive to. Since the use of animals during drug development studies is becoming increasingly difficult, the focus has been on refining, reducing and replacing them. While scientists have been increasingly using human cell lines and other methods, such alternatives have some inherent limitations they cannot mimic the whole organ system. Organoids are a far superior alternative to cell lines.

Organoids offer new opportunities to studying proteins and genes that are critical for the development of an organ. This helps in knowing how a mutation in a specific gene causes a disease or disorder. In a study in Europe using intestinal organoids from six patients with an intestine disorder, it became possible to identify the mutation in a gene that prevented the formation of a healthy intestine. Researchers have used brain organoids to study how the Zika virus affects brain development in the embryo.

Scientists are already using stem cells taken from tumours to grow organoids that are poised to develop cancer. The ability to grow organoids using cancer stem cells allows researchers to study the genes, proteins and signalling pathways that cancer cells use to develop and grow. They are also using healthy organoids to identify and verify the gene mutations that cause cancer.

In an opinion piece in Nature, scientists argued that the largest brain that has been grown in the laboratory is only 4 mm in diameter and contains only 2-3 million cells. In comparison, an adult human brain measures 1,350 cubic centimetres, and has 86 billion neurons and another 86 billion non-neuronal cells and a similar number of non-neuronal cells. The authors argue that organoids do not have sensory inputs and sensory connections from the brain are limited. Isolated regions of the brain cannot communicate with other brain regions or generate motor signals. They wrote: Thus, the possibility of consciousness or other higher-order perceptive properties [such as the ability to feel distress] emerging seems extremely remote.

You have reached your limit for free articles this month.

Register to The Hindu for free and get unlimited access for 30 days.

Find mobile-friendly version of articles from the day's newspaper in one easy-to-read list.

Enjoy reading as many articles as you wish without any limitations.

A select list of articles that match your interests and tastes.

Move smoothly between articles as our pages load instantly.

A one-stop-shop for seeing the latest updates, and managing your preferences.

We brief you on the latest and most important developments, three times a day.

*Our Digital Subscription plans do not currently include the e-paper ,crossword, iPhone, iPad mobile applications and print. Our plans enhance your reading experience.

Here is the original post:
Can organoids, derived from stem cells, be used in disease treatments? - The Hindu

University team to seek approval for iPS-based heart treatment trial – The Japan Times

OSAKA A university research team will seek government approval by the end of October to carry out a clinical trial using iPS cells to treat a serious heart condition, Osaka University officials said Wednesday.

The treatment involves transplanting sheets of heart muscle cells, generated from induced pluripotent stem cells that can develop into any type of tissue, to individuals suffering from ischemic heart disease.

The disease is caused by the buildup of plaque in the coronary arteries, which partially or totally blocks blood flow to the heart.

The team, led by Yoshiki Sawa, a professor at Osaka Universitys Department of Cardiovascular Surgery, received approval for a clinical study from the Ministry of Health, Labor and Welfare in May 2018.

But the study was delayed after a powerful earthquake hit western Japan a month later, damaging a research facility where the necessary cells would have been cultivated.

View original post here:
University team to seek approval for iPS-based heart treatment trial - The Japan Times

Global Gemcitabine Hydrochloride Market: Segmented By Application And Geography Trends, Growth And Forecasts To 2024 – Health News Office

The Induced Pluripotent Stem Cells (IPSCS)market research report added by Report Ocean, is an in-depth analysis of the latest trends, market size, status, upcoming technologies, industry drivers, challenges, regulatory policies, with key company profiles and strategies of players. The research study provides market introduction, INDUCED PLURIPOTENT STEM CELLS (IPSCS) market definition, regional market scope, sales and revenue by region, manufacturing cost analysis, Industrial Chain, market effect factors analysis, INDUCED PLURIPOTENT STEM CELLS (IPSCS) market size forecast, 100+ market data, Tables, Pie Chart, Graphs and Figures, and many more for business intelligence.

UPTO 30% OFF ON SINGLE USER PDF: https://www.reportocean.com/industry-verticals/sample-request?report_id=Global-Induced-Pluripotent-Stem-Cells-(iPSCs)-Market-OWL24961

INDUCED PLURIPOTENT STEM CELLS (IPSCS) Market: Competitive Analysis by Key Players:

The Global INDUCED PLURIPOTENT STEM CELLS (IPSCS) Market also explains the competitive landscape among the major key players of the market, based on various parameters, such as:

INDUCED PLURIPOTENT STEM CELLS (IPSCS) Market Segments:

Market Segment by Manufacturers,> Fujifilm Holding Corporation (CDI)> ReproCELL> Astellas Pharma Inc> Ncardia> Cell Inspire Biotechnology> Sumitomo Dainippon Pharma> Pluricell Biotech> Fate Therapeutics, Inc

Market Segment by Type,> Human iPSCs> Mouse iPSCsMarket Segment by Applications,> Academic Research> Drug Development and Discovery> Toxicity Screening> Regenerative Medicine

Key Developments in the Induced Pluripotent Stem Cells (iPSCs) MarketTo describe Induced Pluripotent Stem Cells (iPSCs) Introduction, product type and application, market overview, market analysis by countries, market opportunities, market risk, market driving force;To analyze the manufacturers of Induced Pluripotent Stem Cells (iPSCs), with profile, main business, news, sales, price, revenue and market share in 2016 and 2018;To display the competitive situation among the top manufacturers in Global, with sales, revenue and market share in 2016 and 2018;To show the market by type and application, with sales, price, revenue, market share and growth rate by type and application, from 2013 to 2019;To analyze the key countries by manufacturers, Type and Application, covering North America, Europe, Asia Pacific, Middle-East and South America, with sales, revenue and market share by manufacturers, types and applications;Induced Pluripotent Stem Cells (iPSCs) market forecast, by countries, type and application, with sales, price, revenue and growth rate forecast, from 2019 to 2026;To analyze the manufacturing cost, key raw materials and manufacturing process etc.To analyze the industrial chain, sourcing strategy and downstream end users (buyers);To describe Induced Pluripotent Stem Cells (iPSCs) sales channel, distributors, traders, dealers etc.To describe Induced Pluripotent Stem Cells (iPSCs) Research Findings and Conclusion, Appendix, methodology and data source

Get a Sample Report for more Expert and Official insights: @ https://www.reportocean.com/industry-verticals/sample-request?report_id=Global-Induced-Pluripotent-Stem-Cells-(iPSCs)-Market-OWL24961

Regional Analysis:

The market research study offers in-depth regional analysis along with the current market scenarios. The major regions analyzed in the study are:

Key highlights and important features of the Report:

Overview and highlights of product and application segments of the global INDUCED PLURIPOTENT STEM CELLS (IPSCS) Market are provided. Highlights of the segmentation study include price, revenue, sales, sales growth rate, and market share by product.

Explore about Sales data of key players of the global INDUCED PLURIPOTENT STEM CELLS (IPSCS) Market as well as some useful information on their business. It talks about the gross margin, price, revenue, products, and their specifications, type, applications, competitors, manufacturing base, and the main business of key players operating in the INDUCED PLURIPOTENT STEM CELLS (IPSCS)Market.

Explore about gross margin, sales, revenue, production, market share, CAGR, and market size by region.

Describe INDUCED PLURIPOTENT STEM CELLS (IPSCS) Market Findings and Conclusion, Appendix, methodology and data source;

Research Methodology:

The market research was done by adopting various tools under the category of primary and secondary research. For primary research, experts and major sources of information have been interviewed from suppliers side and industries, to obtain and verify the data related to the study of the Global INDUCED PLURIPOTENT STEM CELLS (IPSCS)Market. In secondary research methodology, various secondary sources were referred to collect and identify extensive piece of information, such as paid databases, directories and annual reports and databases for commercial study of the GlobalINDUCED PLURIPOTENT STEM CELLS (IPSCS) Market. Moreover, other secondary sources include studying technical papers, news releases, government websites, product literatures, white papers, and other literatures to research the market in detail.

Get in Touch with Us:

Name: David McKee

Email: [emailprotected]

Tel: +1 888 212 3539 (US TOLL FREE)

Website: https://www.reportocean.com/

Excerpt from:
Global Gemcitabine Hydrochloride Market: Segmented By Application And Geography Trends, Growth And Forecasts To 2024 - Health News Office

What is aplastic anemia? Symptoms, causes, and treatment – Medical News Today

Aplastic anemia is a medical condition that damages stem cells in a person's bone marrow. These cells are responsible for making red blood cells, white blood cells, and platelets, which are vital to human health.

Doctors believe various conditions can cause aplastic anemia, while the disease itself ranges in severity from mild to life threatening.

Medical advancements mean that aplastic anemia is more treatable than ever. In this article, learn more about this rare medical disorder.

When a person has aplastic anemia, their bone marrow does not create the blood cells it needs. This causes them to feel ill and increases their risk of getting infections.

Doctors also call aplastic anemia bone marrow failure.

Doctors do not know exactly how many people in the United States have aplastic anemia.

According to the National Organization for Rare Disorders (NORD), doctors diagnose approximately 500 to 1,000 cases every year. It is most common in older children, teenagers, and young adults.

Researchers believe that most cases of aplastic anemia are due to the immune system attacking healthy bone marrow cells, according to NORD.

Doctors have also identified some of the possible causes of this immune system response, including:

However, doctors usually cannot pinpoint the underlying cause in most aplastic anemia cases.

When the cause is unknown, doctors refer to the condition as idiopathic aplastic anemia.

Symptoms of aplastic anemia include:

These symptoms may be severe. Some people may have heart-related symptoms, such as chest pain.

A doctor will start by asking about a person's symptoms and their medical history.

They will usually use a blood test known as a complete blood count (CBC) to evaluate a person's red blood cells, white blood cells, and platelets. If all three of these components are low, a person has pancytopenia.

A doctor may also recommend taking a sample of bone marrow, which comes from a person's pelvis or hip.

A laboratory technician will examine the bone marrow. If a person has aplastic anemia, the bone marrow will not have typical stem cells.

Aplastic anemia can also have similar symptoms as other medical conditions, such as myelodysplastic syndrome and paroxysmal nocturnal hemoglobinuria. A doctor will want to rule out these conditions.

Sometimes, a person with other medical conditions can develop aplastic anemia. These conditions include:

If a person has these conditions, a doctor will recognize that they are more likely to get aplastic anemia.

Doctors usually have two goals when treating aplastic anemia. The first is to reduce the person's symptoms, and the second is to stimulate the bone marrow to create new blood cells.

People with aplastic anemia can receive blood and platelet transfusions to correct low blood counts.

A doctor may also prescribe antibiotics as a person needs white blood cells to fight infections. Ideally, these drugs will prevent infections until a person can build more new white blood cells.

Doctors usually recommend a bone marrow transplant to stimulate new cell growth in the long term.

For this, a doctor may first prescribe chemotherapy medications to kill off abnormal bone marrow cells that are affecting a person's overall bone marrow function.

Next, a doctor performs a bone marrow transplant by injecting the bone marrow into a patient's body.

Ideally, the individual will receive bone marrow from a close family member. However, even a sibling donor is only a match in 2030% of cases.

People can also receive bone marrow from someone who is not related to them if doctors can find a compatible donor.

Some people cannot tolerate bone marrow transplants, especially older adults, and those having difficulty recovering from chemotherapy. Others may not be able to find a donor that matches their bone marrow. In these instances, a doctor can prescribe immunosuppressive therapy.

Immunosuppressive medicines suppress the immune system, which ideally stops it from attacking healthy bone marrow cells. Examples of these medications include antithymocyte globulin (ATG) and cyclosporine.

According to NORD, an estimated one-third of people with aplastic anemia do not respond to immunosuppressive drugs.

If this is the case, doctors may consider other treatments, such as hematopoietic stem cell transplantation and a medication called eltrombopag (Promacta).

Those with aplastic anemia may face complications due to their disease as well as their treatment.

Sometimes, a person's body rejects a bone marrow transplant. Doctors call this graft-versus-host disease or GVHD.

GVHD can make a person feel extremely ill and can cause symptoms that include:

According to 2015 research, about 15% of aplastic anemia patients who receive immunosuppressive therapy will develop myelodysplastic syndromes or acute myeloid leukemia.

These conditions can develop years after a person's initial diagnosis.

Some people do not respond to aplastic anemia treatments. When this is the case, they are more vulnerable to infections that can be life threatening.

The outlook for a person with aplastic anemia depends on many factors, including:

A doctor will discuss a person's treatment outlook when considering the various therapies.

Aplastic anemia damages stem cells in a person's bone marrow. The bone marrow makes red blood cells, white blood cells, and platelets, which are all essential for the body.

A person with aplastic anemia may experience severe anemia symptoms. Treatment may include chemotherapy, stem cell transplants, and immunotherapy.

Read the original post:
What is aplastic anemia? Symptoms, causes, and treatment - Medical News Today

American Academy of Stem Cell Physicians Announced Today That Their Safety Panel Session is Open and Free to the Public – P&T Community

MIAMI, Oct. 24, 2019 /PRNewswire/ -- The AASCP has recently created guidelines thatare current safety recommendations given to physicians who are using biologics in their medical practice. A highly anticipated and sought after Safety StandardsPanel session, hosted by AASCP on Nov. 2, 2019, will be moderated by The Alliance for Cell Therapy Now,with President Ms. Janet Marchbrody.The sessions normally are closed to the public but this particular SafetyStandard Panel discussion will be open to the public, covering the growing safety concerns of the industry.

Alliance for Cell Therapy Now is a coalition of organizations representing patients, health care providers and the academic and scientific community, who are working together to advance safe and effective regenerative cell therapies. The mission is to advance the development, manufacturing and delivery of safe and effective regenerative cell therapies through policy development, consensus and advocacy. Alliance for Cell Therapy Now is bringing together experts and stakeholders to gain consensus on and advocate for policies that will advance the science and the field, including those focused on promoting clinical research, assuring the adoption of consensus standards to promote safety and quality, building capacity and expertise within the workforce, and establishing a national outcomes database to advance the science, promote improvements in quality and safety, and inform regulatory, paymentand patient decision-making.

Alliance for Cell Therapy Now is guided by an Advisory Board comprised of leaders in the scientific, academicand patient communities; Ms.Janet M. MarchibrodaPresident, Alliance for Cell Therapy Now Fellow, Bipartisan Policy Center Senior Vice President, Health Policy, Bockorny Group, has agreed to join theAASCP as a moderator for their SafetyPanelat The Hyatt Regency in Miami. This particular coveted safetypanel session will be open to the public and broadcast live on YouTube at 3:00 p.m. on Nov. 2, 2019.

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. The AASCP also recommends 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, Allografts and Amniotic Fluids and are considered the most experienced leaders for proper advocacy in the field. The AASCP is involved directly with other authorities within the field and seeks only to bring knowledge and awareness for the ever growing regenerative medicine industry.My hope is that the SafetyPanel discussion on Nov.2, 2019, is to help get rid of the bad actors that are damaging the field for everyone."

AASCP is hosting their medical conference in Miami on Nov. 1-3 , 2019. Sessions are normally closed to the public and, therefore, require registration. The conference is taking place at the downtown MiamiHyatt Regency, located at 400 SE 2nd Ave, Miami, FL 33131.Becauseof limited seating, we encourage everyone to please RSVP ataascp.net andto register.

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

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

Related Images

dr-farshchian-teaching-at-aaoscp.jpeg Dr. Farshchian teaching at AAOSCP workshop Dr. Alimorad Farshchian speaking at AASCP

Related Links

AASCP Safety guidelines

AASCP website / registration

View original content to download multimedia:http://www.prnewswire.com/news-releases/american-academy-of-stem-cell-physicians-announced-today-that-their-safety-panel-session-is-open-and-free-to-the-public-300945080.html

SOURCE The American Academy of Stem Cell Physicians

See the original post:
American Academy of Stem Cell Physicians Announced Today That Their Safety Panel Session is Open and Free to the Public - P&T Community