Ozzy Osbourne Recovering from Recent Health Issues and Working on New Album – mxdwn.com

Ariel King July 18th, 2020 - 8:30 PM

Ozzy Osbourne is currently recovering from recent health issues related to Parkinsons, according to his wife, Sharon Osbourne. Ozzy Osbourne is also currently working on a new album.Sharon Osbourne made the comment in an interview on Steve-Os Wild Ride! Sharon Osbourne remarked on Osbournes upcoming album.

Hes starting his second album with Andrew Watt right now, Sharon Osbourne said. And you cant stop him. Hes doing it.

Ozzy Osbournes last album, Ordinary Man, had been released last February, with Andrew Watt producing the album. Ordinary Man had been Osbournes first solo album in ten years. The singer first announced plans to begin working on a new album only a few days after the release of Ordinary Man.

Hes doing really, really good, Sharon Osbourne said. Hes had a terrible, terrible injury. At one point, they thought he would never walk again, but he is hes walking and hes doing great. Hes been hit by so much medically, but hes doing good. Hes getting stronger every day.

Ozzy Osbourne had suffered a fall in 2019, while he had a year left until the completion of his farewell tour. The fall resulted in a spinal injury which resulted in surgery, with Sharon Osbourne saying it sparked off Ozzy Osbournes Parkinsons disease.

Osbourne revealed he had been diagnosed with Parkinsons at the beginning of this year, the singer undergoing stem cell treatments to mitigate the symptoms. His daughter, Kelly Osbourne, revealed in April that the treatment had been working with remarkable results. Shortly after being diagnosed, Ozzy Osbourne had cancelled his last tour, the singer hoping to resume dates prior to the pandemic.

See original here:
Ozzy Osbourne Recovering from Recent Health Issues and Working on New Album - mxdwn.com

What the research says about Covid-19 treatments – ETHealthworld.com

NEW DELHI: There is no cure yet for Covid-19. Doctors and scientists are scrambling to find treatments and drugs that can save the lives of infected people and perhaps prevent infection. Even the most promising treatments to date only help certain groups of patients, and await validation from further trials. Here are 16 major treatments assessed by The New York Times, and what the latest research says about them.

BLOCKING THE VIRUS Antiviral drugs can stop viruses such as HIV and hepatitis C from hijacking our cells. Scientists are searching for antivirals that work against the new coronavirus. Remdesivir (Promising evidence) It stops viruses from replicating by inserting itself into new viral genes. While it didnt fulfill its original purpose of fighting Ebola and hepatitis C, preliminary data suggests it can reduce hospital stay in severe cases from 15 to 11 days. The latest data also hints that it might reduce death rates among those who are very ill.

77027170

Favipiravir (Tentative or mixed evidence) Favipiravir is a flu drug that blocks a viruss ability to copy its genetic material. A small study in March indicated that it might clear out the coronavirus from the airway, but results from larger, well-designed clinical trials are still pending.

EIDD-2801 (Tentative or mixed evidence) Another antiviral originally designed to fight the flu, EIDD-2801 has had promising results against the new coronavirus in studies in cells and on animals. It is still being tested in humans. Recombinant ACE-2 (Tentative or mixed evidence) The coronavirus enters a cell by latching on to a human protein called ACE-2. Recombinant (artificial) ACE-2 proteins might be able to lure it away from vulnerable cells. They have shown promising results in experiments on cells, but not yet in animals or people. Lopinavir-Ritonavir (Not promising) At first, this combination of HIV drugs seemed to stop the new coronavirus from replicating, but clinical trials in patients proved disappointing and the WHO suspended them this month. However, the drugs might still play a role as a preventive and in treating patients with mild symptoms. Hydroxychloroquine and Chloroquine (Not promising) At the start of the pandemic, researchers found that these old anti- malaria drugs could stop the coronavirus from replicating in cells. A few small studies on patients also offered hope that hydroxychloroquine could treat Covid. However, data from randomised clinical trials shows hydroxychloroquine didnt help people with Covid-19 get better or prevent healthy people from contracting the coronavirus. Also, giving hydroxychloroquine to people right after being diagnosed didnt reduce the severity of their disease. The US Food and Drug Administration (FDA) now warns that the drug can cause serious side effects to the heart and other organs when used to treat Covid-19. MIMICKING THE IMMUNE SYSTEM Most patients fight off the virus with a strong immune response. Drugs might help people who cant mount an adequate defense

Convalescant Plasma (Tentative or mixed evidence) A century ago, doctors used plasma from the blood of recovered flu patients to treat people sick with flu. The same strategy has been tried on severely ill Covid patients and the early results are promising. The FDA has authorised plasma therapy for very sick Covid patients.

Monoclonal antibodies (Tentative or mixed evidence) Convalescent plasma contains a mix of antibodies, only some of which can fight the coronavirus. Researchers have identified the most potent Covid antibodies and their synthetic copies known as monoclonal antibodies can be manufactured in bulk and injected into patients. Safety trials for this treatment have only just begun.

Interferons (Tentative or mixed evidence) Interferons are molecules that our cells produce to make the immune system attack viruses. Injecting synthetic interferons is now a standard treatment for immune disorders. Experiments in mice and cells suggest that interferon injections could be used both as a preventive and a treatment for Covid-19.

PUTTING OUT FRIENDLY FIRE The most severe symptoms of Covid-19 are the result of the immune systems overreaction to the virus. Scientists are testing drugs that can rein in its attack

Dexamethasone (Promising evidence) This cheap and widely available steroid blunts many types of im- mune responses. Doctors have long used it to treat allergies, asthma and inflammation. A study of more than 6,000 people found that dexametha- sone reduced deaths by one-third in patients on ventilators, and by one- fifth in patients on oxygen.

Cytokine inhibitors(Tentative or mixed evidence) The body produces molecules called cytokines to fight off diseases. But in excess, they can trigger a cy- tokine storm, which causes the immune system to overreact to in- fections. Researchers have created several drugs to halt cytokine storms, including tocilizumab, sari- lumab and anakinra. While a few have offered modest help in some trials, others have faltered. The drug company Regeneron recently announced that a branded version of sarilumab, Kevzara, failed Phase 3 clinical trials.

Cytosorb (Tentative or mixed evidence) Cytosorb is a cartridge that filters cytokines from the blood in an at tempt to cool cytokine storms. The machine can purify a patients entire blood supply about 70 times in a 24-hour period.

Stem cells (Tentative or mixed evidence) Certain kinds of stem cells can secrete anti-inflammatory molecules. Researchers have tried to use them to treat cytokine storms, and now dozens of clinical trials are under way to see if they can help Covid-19 patients. But stem cell treatments havent worked well in the past, and its not clear yet if theyll work against the coronavirus.

OTHER TREATMENTS Doctors and nurses often administer other supportive treatments to help patients with Covid-19.

Prone positioning (Widely used) The simple act of flipping Covid-19 patients onto their bellies opens up the lungs. The manoeuvre might help some individuals avoid the need for ventilators entirely.

Ventilators (Widely used) Devices that help people breathe are an essential tool. Some patients do well if they get an extra supply of oxygen through the nose or via a mask connected to an oxygen ma- chine. Patients in severe respira- tory distress may need to have a ventilator breathe for them until their lungs heal.

Anticoagulants (Tentative or mixed evidence) The coronavirus can invade cells in the lining of blood vessels, leading to tiny clots that can cause strokes. Anticoagulants are commonly used to slow the formation of clots, and doctors sometimes use them on Covid-19 patients with clots.

Original post:
What the research says about Covid-19 treatments - ETHealthworld.com

Stem Cells for Everyone: Revolutionizing Regenerative …

Induced pluripotent stem cells (iPS cells or iPSCs) are stem cells induced from somatic cells that are reprogrammed to an embryonic stem cell-like state by introducing special factors (genes). iPSCs are able to become any type of cells in the body and proliferate almost indefinitely, like an embryonic stem cell. Unlike embryonic stem cells, iPSCs can be made from matured cells in the body, such as skin or blood cells, from anyone. iPSCs-derived cell therapy generated from a patient's own cells minimizes the risk of immune rejection. It is expected to change the course of regenerative medicine, drug discovery, and personalized medicine.

Unlike other stem cells such as mesenchymal stem cells (MSCs) and hematopoietic stem cells (HSCs), iPSCs can differentiate into all tissue and cell types, can be made with a small amount of cells, and can be grown to quantities necessary. These unique abilities make iPSCs unrivaled as stem cells of choice for patient-specific cell therapy and drug discovery. For example, COVID19/SARS-CoV-2-targeted lung cells, differentiated from patient-derived iPSCs, are a valuable in vitro disease model and can be used for drug and vaccine discovery for SARS-CoV-2.

There are numerous ongoing preclinical and clinical studies involving iPSCs for diseases such as age-related macular degeneration, spinal cord injury, heart failure, GvHD, etc. with several of them yielding positive results. However, the manufacturing of high quality, clinical-grade iPSCs currently faces a bottleneck. The iPSCs used in the first clinical trial in Japan cost approximately one million USD and took one year to generate. At this cost and the rate of production, personalized stem cell-based medicine would not be practical.

I Peace'snovel methodology to manufacture clinical-grade iPSCs in an automated closed, compact, and modular device provides the scalability required formass parallel production of personalized clinical-grade iPSC lines within the I PeaceGMP facility. I Peace will shortly begin gradually increasing its production capability while carefully examining logistical issues associated with mass production of iPSCs. This technology enables dramatic cost reduction and efficient production of clinical-grade iPSCs from multiple donors at the same time,paving the way for a future of global personalized stem cell-based medicine.

Background on the development of the Fully Closed Automatic iPSC Mass Manufacturing System

Existing methods of iPSC generation are labor and cost-intensive, with low efficiency. Clinical-grade iPSC manufacturing requires exclusive use of a whole clean room for just one donor over a long period of time, which meant that mass production was not practical and the associated cleanroom costs were enormous. This is a large barrier in making iPSC-derived medical treatments available to all.

Due to these factors, it was challenging to efficiently mass-produce iPSCs from multiple donors. As a result, only a limited number of clinical-grade iPSC lines were available, with their very high cost as barrier to widespread use, up until now.

Outline of the device

The fully closed automated iPSC manufacturing system that I Peace has successfully developed is different from an automated iPSC culturing system, which simply grows the iPSCs generated elsewhere. Instead, our compact closed-system is capable of reprogrammingcarrying out the full sequence of processes required to change the cell fate of donor cells into iPSCs. The devicecarries no risk of cross-contamination between donors or from outside. Being modular and scalable with a small footprint, many units can be operated in parallel to carry out mass production of clinical-grade iPSCs from a large number of donors simultaneously in a single room. The whole systemfrom the individual biological steps to the overall operationis automated, and the joint development project with FANUC CORPORATIONincluded the creation of an automated operating system using robots.

This technology will revolutionize both allogeneic and personalized regenerative medicine. Unclogging the bottleneck of a limited number of available clinical-grade iPSC lines, this technology will allow us to offerresearchers and institutions a steady supply ofdifferent clinical-grade iPSC lines from which they can select the iPSC line(s) best suited for their particulararea of clinical research. This will be game-changing in accelerating the pace of clinical research using iPSCs. Additionally, the system's ability to simultaneously produce iPSCs from different donors makes personalized medicine possible. The technology will also accelerate drug discovery. Whereas up until now, drug discovery and regenerative medicine research have relied ona limited number of disease-specific iPSC lines, it will now be possible to prepare large libraries of iPSCs from patients and healthy individuals, which we believe will lead to faster discovery of better drugs.

Adopting as its motto 'Peace of mind with iPSCs,' I Peace, Inc. has been working to create a world in which iPSC-based medical treatments are available to everyone. Theclosed-system automated iPSC production device makes iPSCmass productionat dramatically reduced cost possible, which represents a great step forward toward a world where iPSC treatments are available to everyone.

Going forward, the demand for iPSCs is expected to grow further as research progresses into regenerative medicine, new drug development, and a wide variety of other areas where iPSCs are utilized. To meet the iPSC demand expected in areas such as cell therapy, drug discovery research, and clinical trials, I Peace isworking to have the system up and running by the end of 2020. I Peace iscommitted to working towards our vision of a future where each person has their own iPSCs banked for immediate use when necessary.

Supporting Information

Key Takeaways:

About I Peace, Inc.

I Peace, Inc. was founded in 2015at Palo Alto, California. I Peace's mission is to alleviate the suffering of diseased patients and help healthy people maintain a high quality of life. I Peace's proprietary manufacturing platform enables the fully-automated mass production of discrete iPSCs from multiple donors in a single room. Increasing the available number of clinical-grade iPSC lines allows our customers to take differentiation propensity into account to select the most appropriate iPSC line for their clinical research at a significantly reduced cost. Our goal is to give every individual the possibility of their own source of personalized stem cells for life through the creation of iPSCs.

Headquarters: Palo Alto, California Website: https://www.ipeace.com

AboutFounder and CEO Dr. Koji Tanabe

Dr. Koji Tanabe obtained his Ph.D. from Kyoto University Graduate School of Medicine, working in the laboratory of Professor Shinya Yamanaka, 2012 Nobel Prize Winner in Physiology/Medicine. There, he spent eight years researching iPS cells starting in 2006 the early days of iPSC development and became the second author of the scientific paper reporting the world's first successful generation of human iPSCs. After getting his Ph.D., Dr. Tanabe moved to the United States and joined the Dr. Marius Wernig Laboratory, part of the Institute for Stem Cell Biology and Regenerative Medicine at Stanford University Medical School, where the world's first successful direct reprogramming from skin cells to neural cells was achieved. Dr. Tanabe's post-doctoral work at Dr. Wernig's lab was on direct reprogramming of blood cells to neural cells and the iPSC reprogramming mechanism, where he also contributed to numerous scientific papers on iPSCs and on direct reprogramming to neural cells. After a period as a guest researcher at Stanford, Dr. Tanabe assumed his present position as CEO of I Peace. He has been awarded an Overseas Research Fellowship by the Japan Society for the Promotion of Science.

SOURCE I Peace, Inc.

Home

More:
Stem Cells for Everyone: Revolutionizing Regenerative ...

Mesenchymal Stem Cells for Regenerative Medicine for …

1. Introduction

Duchenne muscular dystrophy (DMD) is an X-linked progressive muscle wasting disorder caused by mutations in the DMD gene [1, 2], affecting 1 in 35005000 male births. Serum creatine kinase (CK) levels are elevated at birth, and motor milestones are delayed. Reduced motor skills between age 3 and 5years provoke diagnostic evaluation. Quality of life for boys with DMD is further affected early in life, with the inability to keep up with peers of early school age and loss of ambulation by 12years of age; premature death occurs at 2030years of age due to respiratory and cardiac complications (https://www.duchenne.com/about-duchenne;https://ghr.nlm.nih.gov/condition/duchenne-and-becker-muscular-dystrophy).

Mutations of the DMD gene cause complete (Duchenne) or partial (Becker) loss of dystrophin protein at the sarcolemma [3]. In normal muscle cells, dystrophin forms a complex with glycoproteins at the sarcolemma, forming a critical link between the extracellular matrix (ECM) and the cytoskeleton [4]. Without the complex, the sarcolemma becomes fragile and is easily disrupted by mechanical stress [4, 5].

Except for corticosteroids, there is currently no effective treatment for DMD [7]. In this chapter, we discuss the potential of mesenchymal stem cells as a therapeutic tool for DMD patients. Many researchers prefer the term mesenchymal stromal cells or mesenchymal progenitors to mesenchymal stem cells because mesenchymal stem cells with self-renewal and trilineage differentiation potential are a minor subpopulation in tissue-derived primary cultures of mesenchymal cells. In this chapter, however, we uniformly refer to them as mesenchymal stem cells.

The absence of dystrophin causes loss of the dystrophin-associated protein complex (DAPC) at the sarcolemma. The sarcolemma lacking the complex becomes vulnerable to mechanical stress. In addition, signalling through dystrophin-DAPC-associated molecules such as nNOS is disturbed [4, 5]. As a result, myofibres die in large numbers by contraction-induced mechanical stress, and to regenerate injured myofibres, inflammatory cells begin to remove debris of the muscle tissue; at the same time, muscle satellite cells are activated, proliferate and fuse with damaged myofibres. In the case of DMD, however, the cycle of degeneration and regeneration of myofibres repeats throughout life. Therefore, secondary pathological changes gradually develop, including perturbation of calcium homeostasis, activation of Ca2+-dependent proteases, mitochondrial dysfunction in myofibres, impaired regeneration of myofibres due to exhaustion of satellite cells, prolonged inflammation, disturbed immune response, fibrosis and fatty infiltration, with poor vascular adaptation and functional ischaemia [7]. These secondary pathological changes accelerate the disease course of DMD, resulting in severe loss of myofibres and muscle atrophy. Therefore, in addition to the restoration of dystrophin protein by gene therapy or stem cell therapy, blockage of secondary pathological events is an important therapeutic strategy for DMD (Figure 1).

Deficiency of dystrophin protein at the sarcolemma causes multiple pathological changes in DMD muscle [6, 7].

Upon injury, muscle satellite cells are activated, proliferate, and either fuse with damaged myofibres or fuse with each other to form new myofibres [8]. In DMD muscle, satellite cells compensate for muscle fibre loss in the early stages of the disease but eventually are exhausted. As a result, in DMD muscle, the myofibres are gradually replaced with fibrous and fatty connective tissue. Therefore, stem cell transplantation is expected to be a potential therapy for DMD [9].

There are different kinds of stem cells with myogenic potential in skeletal muscle. Muscle satellite cells are authentic unipotent skeletal muscle-specific stem cells [8]. Muscle-derived stem cells (MDSCs) [10] and mesangioblasts [11] were reported to be multipotent and transplantable via circulation; therefore, they are expected to be promising tools for cell-based therapies for DMD. Recently, muscle progenitors were induced from pluripotent stem cells as a cell source for cell-based therapy of DMD because induced pluripotent stem cells (iPSCs) can be expanded without losing pluripotency [12]. Myogenic cells induced from iPSCs are usually at a foetal stage and poorly engraft in the muscle of immunodeficient DMD model mice [13, 14].

In addition, muscles affected by muscular dystrophies are in a state of continuous inflammation and are characterised by marked and sustained infiltration of inflammatory and immune cells with fibrosis and adipose replacement. Such pathological microenvironments would not support survival, proliferation, and differentiation of the transplanted stem cells. Therefore, researchers have started to consider not only the properties of stem cells but also the microenvironment.

Skeletal muscle regenerates when it is injured. The regeneration process is complex but well organised, depending on the interaction among different types of cells: muscle stem/progenitor cells, muscle-resident mesenchymal progenitors and cells involved in inflammatory and innate and adaptive immune responses. Dynamic extracellular matrix (ECM) remodelling is also required for successful muscle regeneration. In the case of a minor traumatic injury, muscle regeneration is rapidly completed by the interplay of these cells. In muscular dystrophies, however, the degeneration/regeneration process is repeated for a long time, causing exhaustion of muscle satellite cells and finally resulting in severe atrophy of skeletal muscles with a loss of myofibres and extensive fibrosis and fat deposition [15].

Fibro/adipogenic progenitors (FAPs) are tissue-resident mesenchymal stem (or stromal or progenitor) cells [16, 17]. Recently, the necessity of FAPs for skeletal muscle regeneration and maintenance was demonstrated using mouse models [18]. The authors demonstrated that depletion of FAPs resulted in loss of expansion of muscle stem cells (MuSCs) and haematopoietic cells after injury and impaired skeletal muscle regeneration [18]. Furthermore, FAP-depleted mice under homeostatic conditions exhibited muscle atrophy and a loss of MuSCs, revealing that FAPs are essential for long-term homeostatic maintenance of skeletal muscle and the MuSC pool [18].

FAPs have dual functions [19, 20]. In small-scale traumatic muscle injury, they are activated, expand and promote muscle regeneration. When regeneration is completed, FAPs are cleared from the regenerated muscle. In pathological conditions, such as muscular dystrophies, they continue to proliferate and contribute to fibrosis and fatty tissue accumulation.

How is the fate of FAPs regulated? Apparently, FAPs are regulated by signals from myogenic cells and immune cells. Altered signals from these cells in dystrophic muscle change the pro-regenerative FAPs to fibrotic and adipogenic types. Recently, Hogarth et al. reported that annexin A2 accumulation in the myofibre matrix promotes adipogenic replacement of FAPs in dysferlin-deficient LGMD2B model mice. The authors also showed that an MMP-14 inhibitor, Batimastat, inhibited adipogenesis of FAP. The authors speculate that Annexin A2 and MMP-14 both prolong the inflammatory environment, therefore causing excessive expansion of FAP in diseased muscle [21]. Pharmacological inhibition of FAP expansion may be a good strategy to prevent fibro/adipogenic changes in dystrophic muscles.

The signals that regulate FAPs remain largely unclear. Interestingly, treating FAPs of young mdx mice with trichostatin A (TSA), a histone deacetylase inhibitor, blocked their fibrotic and adipogenic differentiation and promoted a myogenic fate [22] by changing chromatin structure [23]. TSA treatment decreased the expression of adipogenic genes and upregulated myogenic genes in FAPs [22].

Inflammatory and immune cells (neutrophils, eosinophils, basophils, macrophage NK cells, dendritic cells, T cells, B cells, etc.) are key regulators of muscle regeneration. In particular, macrophages orchestrate the regeneration process. In the early phase of muscle regeneration, M1 (inflammatory) macrophages remove necrotic tissues by phagocytosis and inhibit fusion of myogenic precursor cells. In the later stage, M2 (regulatory) macrophages gradually replace M1 macrophages and play anti-inflammatory and pro-regenerating roles by promoting the differentiation of myogenic cells and the neovascularization of regenerating muscle regeneration [24].

DMD muscle, which remains dystrophin-deficient, experiences continuous cycles of necrosis and regeneration of myofibres. This causes chronic inflammation and evokes T cell-mediated immune responses, which involves the coexistence of both M1 and M2 macrophages and T cells in the muscle, and it further damages myofibres and exacerbates fibrosis and adipocyte infiltration [6, 25, 26]. Therefore, pharmacological inhibition of excess inflammation and immune response is a reasonable therapeutic strategy for DMD.

As a therapeutic tool for regenerative medicine, mesenchymal stem cells (MSCs) have received significant attention in the recent years due to their high growth potential, paracrine effects, immunomodulatory function and few reported adverse effects [27, 28]. Since MSCs show relatively low immunogenicity due to low expression of major histocompatibility (MHC) antigens and their immunomodulation function, they are being used even in allogeneic settings.

To facilitate research on MSCs, the International Society of Cellular Therapy (ISCT) formulated minimal criteria for defining multipotent MSCs in 2006 [29]. First, MSCs must be plastic adherent when maintained in standard culture conditions. Second, MSCs must express CD105, CD73 and CD90 and must not express CD45, CD34, CD14, CD11b, CD79alpha, CD19 and HLA-DR surface molecules. Third, MSCs must differentiate into osteoblasts, adipocytes and chondrocytes under standard in vitro differentiation protocols [29].

Historically, MSCs were isolated from bone marrow [30, 31, 32, 33]. Currently, MSCs are shown to exist in the perivascular niche in nearly all tissues and are prepared from a variety of tissues, such as the umbilical cord [34], placenta [35], adipose tissue [36] and dental tissues [37]. Preparation of MSCs from those tissues is less invasive than it is from BM. MSCs from different tissues have similar functions, but detailed comparative studies revealed that MSCs of different origins possess different properties [38].

MSCs are multipotent stem cells that undergo self-renewal and differentiate into multiple tissues of the mesenchymal lineage and into a non-mesenchymal lineage, including neurons, glia, endothelial cells, hepatocytes and cells in the pancreas [27]. This wide range of differentiation capacities is one reason why mesenchymal stem cells are being tested in almost 1000 clinical trials in regenerative medicine for the musculoskeletal system, nervous system, myocardium, liver, skin and immune diseases (http://ClinicalTrial.gov). Importantly, the differentiation potential of MSCs varies according to their origin, method of isolation and in vitro propagation procedures [39, 40, 41].

MSCs secrete a variety of bioactive molecules, such as growth factors, chemokines and cytokines. These molecules regulate the survival, proliferation and differentiation of target cells, promote angiogenesis and tissue repair and modulate inflammation and innate or acquired immunity. It is widely accepted that the therapeutic effects of MSCs in preclinical and clinical trials are largely due to their paracrine function [27]. Importantly, the secretome of MSCs varies depending on the age of the donor and the niches where the cells reside [42]. Therefore, it is expected that the therapeutic effects of MSCs with different origins exert will be different.

Recently, there has been considerable interest in the clinical application of MSCs for the treatment of muscle diseases. However, the myogenic potential of MSCs is controversial.

Sassoli et al. found that myoblast proliferation was greatly enhanced in coculture with bone marrow MSCs [43]. Myoblasts after coculture expressed higher levels of Notch-1, a key determinant of myoblast activation and proliferation. Interestingly, the effects were mediated by vascular endothelial growth factor (VEGF) secreted by MSCs [43]. A VEGFR2 inhibitor, KRN633, inhibited the positive effects of MSC-CM on C2C12 cell growth and Notch-1 signalling [43]. Linard et al. showed successful regeneration of rump muscle by local transplantation of bone marrow MSCs (BM-MSCs) after severe radiation burn using a pig model [44]. The authors speculate that locally injected BM-MSCs secreted growth factors such as VEGF and promoted angiogenesis. The authors also showed that MSCs supported the maintenance of the satellite cell pool and created a good macrophage M1/M2 balance. Nakamura et al. reported that transplantation of MSCs promoted the regeneration of skeletal muscle in a rat injury model without differentiation into skeletal myofibres. The report suggests that MSCs contribute to the regeneration of skeletal muscle by paracrine mechanisms [45]. Maeda et al. reported that BM-MSCs transplanted into peritoneal cavities of dystrophin/utrophin double-knockout (dko) mice strongly suppressed dystrophic pathology and extended the lifespan of treated mice [46]. The authors speculated that CXCL12 and osteopontin from BM-MSCs improved muscle regeneration. Bougl et al. also reported that human adipose-derived MSCs improved the muscle phenotype of DMD mice via the paracrine effects of MSCs [47].

In addition to soluble factors, recent studies demonstrated that MSCs secrete a large number of exosomes for intercellular communication [48, 49]. These exosomes are now expected to be a therapeutic tool for many diseases [50, 51]. Nakamura et al. reported that exosomes from MSCs contained miRNAs that promoted muscle regeneration and reduced the fibrotic area [45]. Bier et al. reported that intramuscular transplantation of PL-MSCs in mdx mice decreased the serum CK level, reduced fibrosis in the diaphragm and cardiac muscles and inhibited inflammation, partly via exosomal miR-29c [49]. Thus, MSC exosomes or MSC cytokines may provide a cell-free therapeutic strategy as an alternative to transplanting MSCs.

On the other hand, Saito et al. reported that BM-MSCs and periosteum MSCs differentiated into myofibres and restored dystrophin expression in mdx mice, although the efficiency was low (3%) [52]. Liu et al. showed that FLK-1+ adipose-derived MSCs restored dystrophin expression in mdx mice [53]. Feng et al. reported that intravenously delivered BM-MSCs increased dystrophin expression in mdx mice [54]. Vieira et al. reported that intravenously injected human adipose-derived MSCs successfully reached the muscle of golden retriever muscular dystrophy (GRMD) dogs and that they expressed human dystrophin [55]. Furthermore, Park et al. reported that human tonsil-derived MSCs (T-MSCs) differentiated into myogenic cells in vitro, and transplantation promoted the recovery of muscle function, as demonstrated by gait assessment (footprint analysis); furthermore, such treatment restored the shape of skeletal muscle in mice with a partial myectomy of the gastrocnemius muscle [56]. These reports suggest that MSCs directly contribute to the regeneration of myofibres and restore dystrophin expression.

In response to damage signals, perivascular MSCs are activated and recruit inflammatory and immune cells and promote inflammation. At a later stage, MSCs begin to suppress inflammation and the immune response. On the other hand, MSCs in circulation are reported to selectively home towards damaged tissue [57]. Once homed, the inflammatory environment stimulates MSCs to produce a large amount of bioactive molecules or to directly interact with inflammatory and immune cells to regulate inflammation and the immune response.

The therapeutic effects of MSCs in preclinical or clinical trials are thought to be partly the result of modulation of innate and adaptive immunity [27], especially through monocyte/macrophage modulation [28]. Inflammation and immune response are part of the pathology of DMD muscle. Therefore, the immunomodulatory functions of MSCs might be useful for the treatment of DMD.

MSCs are supposed to modulate inflammation and the immune response by (a) suppressing the maturation and function of dendritic cells [58, 59, 60], (b) promoting macrophage differentiation towards an M2-like phenotype with high tissue remodelling potential and anti-inflammatory activity [61], (c) inhibiting Th17 generation and function [62, 63], (d) inhibiting Th1 cell generation [64], (e) suppressing NK [65, 66] and T cytotoxic cell function [66], (f) stimulating the generation of Th2 cells [67] and (g) inducing Treg cells [64, 66, 68].

Pinheiro et al. investigated the effects of adipose-derived mesenchymal stem cell (AD-MSC) transplantation on dystrophin-deficient mice. Local injection of AD-MSCs improved histological phenotypes and muscle function [69]. AD-MSCs decreased the muscle content of TNF-, IL-6, TGF-1 and oxidative stress but increased the levels of VEGF, IL-10 and IL-4 [69]. MSC-derived IL-4 and IL-10 are reported to convert M1 (pro-inflammatory) macrophages to the M2 (anti-inflammatory) type and promote satellite cell differentiation [70]. These results suggest that transplanted AD-MSCs ameliorated the dystrophic phenotype partly by modulating inflammation.

In a clinical trial of gene therapy using a dystrophin transgene, T cells specific to epitopes of pre-existing dystrophin in revertant fibres were detected, suggesting the existence of autoreactive T-cell immunity against dystrophin before treatment [71]. Currently, exon skipping therapy to restore the reading frame of the DMD gene, and readthrough therapy of premature stop codons (e.g. aminoglycosides or ataluren), is being tested in patients with DMD. The treated patients start to produce dystrophin, which provides new epitopes to them. Suppression of undesirable immune responses against newly produced dystrophin might improve the efficiency of gene therapy.

Transplantation of myogenic cells also evokes innate and acquired immune responses against transplanted cells in the recipient. Therefore, immunosuppression by MSCs is expected to improve the engraftment of transplanted cells and the therapeutic effects of cell therapy. In addition, MSCs support the survival, proliferation, migration and differentiation of myogenic cells by secreting trophic factors.

Although BM-MSCs are well studied and widely tested in regenerative medicine, the collection procedure for bone marrow is invasive and painful. In addition, adult BM-MSCs cannot be expanded in culture beyond 10 passages [72]. To obtain MSCs with higher proliferative potential, other sources of MSCs are gaining attention, such as the umbilical cord and the placenta. MSCs from these sources proliferate better than BM-MSCs but still show limited proliferative activity [38].

hiPSCs can be expanded in vitro without loss of pluripotency and are therefore an ideal source for deriving mesenchymal stem cells of high quality in a large quantity [73, 74, 75]. In addition, unlike human ES cells, iPSCs are not accompanied by ethical concerns. To date, many protocols have been reported for the deviation of mesenchymal stem cells from human ES cells/iPS cells [73, 74, 75, 76, 77], although the difference in properties among iMSCs induced by different protocols remains to be determined [73, 74, 77]. For clinical use, iMSCs would be generated from well-characterised, pathogen-free, banked iPSCs with known HLA types or from patient-specific iPSCs.

MSCs induced from human iPS cells are generally characterised as reprogrammed, rejuvenated MSCs with high proliferative activity [78]. A previous study reported that MSCs from human iPSCs could be expanded for approximately 40 passages (120 population doublings) without obvious loss of plasticity or onset of replicative senescence [79]. In addition, iMSCs have been shown to exhibit potent immune-modulatory function and therapeutic properties (Table 1) [80]. Spitzhorn et al. reported that iMSCs did not form tumours after transplantation into the liver [81], but to exclude residual undifferentiated iPS cells, purification of MSCs by FACS using MSC markers and careful evaluation of the risk of tumour formation would be required for each preparation.

Comparison of properties of human iMSCs with human BM-MSCs.

The therapeutic potential of iMSCs has been tested in bone regeneration [80, 84], intestinal healing [85], myocardial disorders [86, 87], limb ischaemia [79] and autoimmune disease [88, 89]. In these studies, iMSCs showed therapeutic effects that were comparable or superior to those of tissue MSCs. In the muscular dystrophy field, there are only a small number of reports so far. Jeong et al. reported that iMSCs transplanted into the tibialis anterior of mdx mice decreased oxidative damage, as evidenced by a reduction in nitrotyrosine levels, and achieved normal dystrophin expression levels [90]. Since direct differentiation of MSCs into myogenic cells is generally limited, the observed effects of iMSCs might be due to the secretion of bioactive molecules that exert immunomodulatory effects and provide trophic support to myogenic cells.

Importantly, however, Liu et al. recently reported that transplantation of BM-MSCs from C57BL/6 mice aggravated inflammation, oxidative stress and fibrosis and impaired regeneration of contusion-injured C57/Bl6 muscle [91]. Although the mechanisms are not clear, the microenvironment in contusion-damaged muscle might induce the transformation of MSCs into the fibrotic phenotype. Caution might be warranted in the clinical application of MSCs to highly fibrotic muscle.

MSCs are multifunctional cells. MSCs secrete trophic factors that help regenerate myofibres. In addition, MSCs suppress inflammation and the immune response in dystrophic mice to protect muscle. MSCs are also expected to support the engraftment of transplanted myogenic cells in recipient muscle. Fortunately, recent technology gives us an option to derive MSC-like cells from pluripotent stem cells. Thus, MSCs are a promising next-generation tool for cell-based therapy of DMD (Figure 2).

Mesenchymal stem cells ameliorate the dystrophic phenotype of DMD muscle. Mesenchymal stem-like cells can be derived from human iPSCs (iMSCs). MSCs, which arrive in the muscle either through direction transplantation or via circulation, secrete a variety of bioactive molecules that promote angiogenesis and support the proliferation and differentiation of satellite cells, thereby promoting muscle regeneration. MSCs also suppress excess inflammatory and immune responses. Whether transplanted MSCs can directly modulate the phenotype of FAPs (resident MSCs) to inhibit fibrosis and fatty replacement remains to be determined. Abbreviations: DC, dendritic cells; NK, natural killer cells; Neu, neutrophil; M, macrophage; T, T lymphocytes; B, B lymphocyte.

A.E. is supported by the Channel System Program (CPS) of the Egyptian and Japanese governments. This study was supported by (1) Research on refractory musculoskeletal diseases using disease-specific induced pluripotent stem (iPS) cells from the Research Center Network for Realization of Regenerative Medicine, Japan Agency for Medical Research and Development (AMED), (2) Grants-in-aid for Scientific Research (C) (16K08725 and 19K075190001) from the Ministry of Education, Culture, Sports, Science and Technology (MEXT), Japan and (3) Intramural Research Grants (30-9) for Neurological and Psychiatric Disorders of NCNP.

The authors declare no conflicts of interest.

Read the original post:
Mesenchymal Stem Cells for Regenerative Medicine for ...

RoosterBio Regenerative Human Bone Marrow Stem Cell …

In the medical field, there have been numerous attempts to get regenerative medicine to work. Regenerative medicine is a branch of medication that deals with the research into the processes involved in replacing or regenerating of the human cells, organs, or tissues.

The effect of this is the reestablishment of the original order of the human body systems. It is a field that holds a lot of promise to engineer damaged or worn out tissues by stimulating the body to repair these tissues.

Regenerative medicine is a branch that also includes growing of the same tissues and organs in a laboratory and implanting them in a patient whose body cannot regenerate the organs by itself.

It is a branch of medication that holds the potential to solve the issue of tissue shortage and also the issue of rejected organs during a transplant.

It is this branch of medication that RoosterBio has involved itself in. The passion of the company is to increase the empowerment of cures that could potentially save lives. They aim to do the same by providing platforms for stem cells that will allow rapid commercial and clinical translation. This will have the effect of rapidly increasing the cell based bio-economy.

The team is comprised of experts that have dedicated themselves in the success of their customers. The staff is also one that is committed to reimagining what the future of stem cells could be. The team is highly responsible and respectful, and works together as a unit to achieve the best results possible.

The company also borrows heavily form the advice of experts. They often consult with the experts in the world of regenerative medicine to ensure that all their products ascribe to the highest standards possible.

The company produces tissues and stem cells that are derived from the human bone marrow and also the human adipose tissue. These cells are specifically known as mesenchymal stem cells and multipotent stem cells respectively.

The company is also highly involved in the production of formulations that are highly engineered to promote the expansion of the same cells.

The company produces the highest volume HMCS. These HMCS are offered by the company in a large variety of formats. The purpose of these cells is to speed up the process of development of the stem cells.

Every vial of cells that you take comes with a guarantee. A guarantee that it will expand 10-fold within a week if you pair it up with the media systems provided by the company.

The media that are sold by the company are designed to accelerate the rapid expansion of mesenchymal cells. The media contain a low volume of very potent serum and also a cocktail of growth factors that are known to mitigate HMSCs. The media from the company is custom built to give you a rapid proliferation rate. This will result in high cell volumes within a brief period of time.

The company also provides you with a donor screening kit. These kits are plug and play and are not complicated. They, therefore, have the effect of saving you a lot of money. The company works with you hand-in-hand to increase the development of your product all the way to the clinic. The purpose of all this is to save you time and money.

The company also has top of the range Clinical Control bioprocess media. These are designed for the 2D batch and 3D fed-batch. They also facilitate easy translation between platforms with the effect of standardizing the results.

The company provides the researchers with the cells at a much larger volume. This means that the researchers can conduct their research at a much faster rate. This is as compared to other companies that only provide the cultures in small quantities. The cells produced by the company are also quite affordable, thereby enabling the researchers to purchase numerous vials.

The customers of the company benefit from both their expertise in manufacturing of stem cells as well as benefitting from the economies of scale offered by the company.

There are no foreseeable downsides, only that the abundance of the cells could raise some flags.

RoosterBio has the aim of providing the researchers into regenerative medicine with enough material to do their research. This availability of material could potentially change the medical world.

[vc_btn title=Click Here To See Our #1 Recommended Omega 3 Fish Oil style=3d shape=square color=juicy-pink size=lg align=center i_icon_fontawesome=fa fa-info-circle link=url:https%3A%2F%2Fsupplementpolice.com%2Ftop-omega-fish-oil|title:Our%20#1%20Recommended%20Fish%20Oil|target:%20_blank button_block=true add_icon=true]

Read this article:
RoosterBio Regenerative Human Bone Marrow Stem Cell ...

3D Cell Culture Market Is Booming Worldwide, Industry analysis with Leading Players Thermo Fisher Scientific, Reprocell Incorporated, Kuraray Co,…

The3D Cell Culture Marketresearch Report is a valuable supply of perceptive information for business strategists. This Premium Tyres Market study provides comprehensive data which enhances the understanding, scope and application of this report.

The key market segments along with its subtypes are provided in the report. This report especially focuses on the dynamic view of the market, which can help to manage the outline of the industries. Several analysis tools and standard procedures help to demonstrate the role of different domains in market. The study estimates the factors that are boosting the development of 3D Cell Culture companies.

You can get the sample copy of this report now @https://www.reportsintellect.com/sample-request/896482

Key Companies Covered : Thermo Fisher Scientific, Reprocell Incorporated, Kuraray Co, Corning, N3d Bioscience, Lonza Group, Insphero, Merck Kgaa, 3D Biotek

You get the detailed analysis of the current market scenario for 3D Cell Culture and a market forecast till 2025 with this report. The forecast is also supported with the elements affecting the market dynamics for the forecast period. This report also details the information related to geographic trends, competitive scenarios and opportunities in the 3D Cell Culture market. The report is also equipped with SWOT analysis and value chain for the companies which are profiled in this report.

Most Important Types : Scaffold-based, Scaffold-free

Most Important Application : Cancer Research, Stem Cell Research, Drug Discovery, Regererative Medicine

Get Instant discount @https://www.reportsintellect.com/discount-request/896482

Global 3D Cell Culture Market Size, Status and Forecast 2020 2025

1 Market Overview

2 Manufacturers Profiles

3 Global 3D Cell Culture Sales, Revenue, Market Share and Competition by Manufacturer

4 Global 3D Cell Culture Market Analysis by Regions

5 North America 3D Cell Culture by Countries

6 Europe 3D Cell Culture by Countries

7 Asia-Pacific 3D Cell Culture by Countries

8 South America 3D Cell Culture by Countries

9 Middle East and Africa 3D Cell Culture by Countries

10 Global 3D Cell Culture Market Segment by Type

11 Global 3D Cell Culture Market Segment by Application

12 3D Cell Culture Market Forecast

13 Sales Channel, Distributors, Traders and Dealers

14 Research Findings and Conclusion

15 Appendixes

Reasons for Buying this Report:

About Us:

Reports Intellect is your one-stop solution for everything related to market research and market intelligence. We understand the importance of market intelligence and its need in todays competitive world.

Our professional team works hard to fetch the most authentic research reports backed with impeccable data figures which guarantee outstanding results every time for you.

So whether it is the latest report from the researchers or a custom requirement, our team is here to help you in the best possible way.

Contact Us:

[emailprotected]

PH +1-706-996-2486

US Address:

225 Peachtree Street NE,

Suite 400,

Atlanta, GA 30303

Visit link:
3D Cell Culture Market Is Booming Worldwide, Industry analysis with Leading Players Thermo Fisher Scientific, Reprocell Incorporated, Kuraray Co,...

Newest Clinic Opens Today at The Villages in Central …

SUNRISE, FL April 9, 2018 U.S. Stem Cell, Inc. (OTC: USRM), a leader in the development of proprietary, physician-based stem cell therapies and novel regenerative medicine solutions, today announced the opening its first USRM clinic in North Central Florida.

The U.S. Stem Cell Clinic of The Villages will offer alternatives to chronic pain management using USRMs proprietary AdipoCell TM product in a minimally invasive procedure utilizing a patients own (autologous) stem cells. Medical director for the clinic will be Dr. Rosemary Daly, an interventional spine/pain management physician who is board certified in Anesthesiology and Pain Medicine. Dr. Daly, who graduated from the New York College of Osteopathic Medicine, is also medical director for the Regenerative Wellness Clinic (RWC) in West Palm Beach, which is also in the USRM network of stem cell treatment centers.

Expansion of our regenerative healing centers to The Villages is an opportunity to directly serve an active community that is very proactive about seeking ways to increase their quality of life, said Dr. Kristin Comella, USRMs Chief Science Officer. We look forward to serving this vibrant community by offering holistic stem cell therapy for neurological, autoimmune, orthopedic and degenerative conditions.

Our vision at USRM has always been to expand access of autologous stem cell treatments to as many people as possible, said Mike Tomas, President and CEO of U.S. Stem Cell, Inc. Now that we are seeing a steady increase in demand for stem cell therapy, which has driven our progress in reaching milestones that allow for growth, we are ready to continue expanding our network of clinics. The clinic at The Villages represents that next step.

USRM has been instrumental in performing more than 10,000 stem cell procedures in the past 19 years for a variety of indications. USRM also trains and certifies physicians in stem cell therapy to date, more than 700+ physicians worldwide and has a growing network of 288 physicians and clinics utilizing the USRM technologies, protocols and products. Dr. Comella continues to enhance USRMs visibility worldwide for autologous stem cell treatments, as well as developing and bringing USRMs proprietary Adipocell TM product to market: a stem cell kit which enables physicians to separate potent stem cells from a patients own fat cells, which are harvested and reinserted in a minimally invasive, two-hour procedure without general anesthesia.

Dr. Comella is also well published in the scientific literature and has been recognized by her peers as an innovator and world leader in the development and clinical practice of stem cell products and therapies. Most recently, she presented a summary of peer-reviewed publications at the Academy of Regenerative Practices Conference in Weston, Florida.

U.S. Stem Cells novel advancements in stem cell therapy, in managements opinion, could be a significant alternative to the explosive opioid crisis in America a direct results of Americas failed attempt at pain management. According to The New York Times, drug overdose is now the leading cause of death in the United States for patients under the age of 50, while the October 15, 2017, edition of 60 Minutes reports that the opioid crisis has claimed 200,000 lives in the United States over the past few decades.

U.S. Stem Cell, Inc. is an emerging leader in the regenerative medicine / cellular therapy industry specializing in physician training and certification and stem cell products including its lead product Adipocell TM , as well as veterinary stem cell training and stem cell banking and creation and management of stem cell clinics. To managements knowledge, USRM has completed more clinical treatments than any other stem cell company in the world.

Read the original:
Newest Clinic Opens Today at The Villages in Central ...

The Play On Omeros – Seeking Alpha

Without education, we are in a horrible and deadly danger of taking educated people seriously." G.K. Chesterton

Today, we revisit a name we have covered often over the past two years. The company has been maddening for investors as the shares trade at the same level as a year ago even as the company continues to push forward its main development asset. We update our investment thesis on this 'Tier 3' concern and outline our investment strategy around its stock in the paragraphs below.

Omeros (OMER) is a Seattle, Washington-based biopharmaceutical company that IPO'd in 2009. The firm is somewhat unique in that it's not focusing on a single therapeutic area or drug platform, rather the company is looking to build a robust, diverse pipeline of first-in-class small-molecule and antibody therapeutics. The company is developing drugs for orphan indications, large-market, and where there's no approved treatments. Omeros currently has one approved therapeutic on the market called Omidria. The drug is designed to prevent miosis in cataract surgery and to reduce postoperative pain. Furthermore, the drug has demonstrated the ability to reduce complications, to prevent intraoperative floppy iris syndrome, and to reduce the need for opioids, pupil-expansion devices and postoperative steroids.

The company's pipeline is currently comprised of 7 product candidates, most of which are in the early stages of development. The lead product candidate is called Narsoplimab, which is being tested in a variety of indications. Omeros has a market capitalization of roughly $800 million and trades for around $14.50 a share.

Pipeline

Narsoplimab

Narsoplimab is a human monoclonal antibody targeting mannan-binding lectin-associated serine protease-2, aka MASP-2. MASP-2 is the effector enzyme of the lectin pathway of the complement system. The lectin pathway is one of the principal pathways of the complement system and is triggered mainly by tissue damage and microbial infection. Critically, inhibition of MASP-2 does not affect the classical complement pathway, which is a critical component of the immune response to infection. This means that the drug can prevent complement-mediated inflammation and endothelial damage while leaving entirely intact the important functions of the other pathways of innate immunity. The drug is being evaluated in a variety of indications: hematopoietic stem cell transplant-associated TMA, IgA nephropathy, atypical hemolytic uremic syndrome, and lupus nephritis/other renal diseases. In the United States, Narsoplimab has been granted FDA Breakthrough Therapy designation in patients who have high-risk HSCT-TMA, and for igA nephropathy; Orphan Drug designation for the prevention of complement-mediated TMAs, for the treatment of HSCT-TMA, and for igA nephropathy; and the drug has received FDA Fast Track designation for the treatment of aHUS. In the European Union, Narsoplimab has been designated an Orphan Medicinal Product for treatment in hematopoietic stem cell transplantation, and for IgA nephropathy. Recently, the company submitted the second part of their rolling BLA for Narsoplimab for the treatment of hematopoietic stem cell transplant-associated thrombotic microangiopathy. The submission was comprised of information on the chemistry, manufacturing and controls of Narsoplimab.

Source: Company Presentation

On March 3rd, the company reported clinical data from their pivotal trial of Narsoplimab for the treatment of hematopoietic stem cell transplant-associated thrombotic microangiopathy. The drug significantly exceeded the FDA-agreed upon threshold for the primary efficacy endpoint of a 15% complete response rate. Furthermore, the drug was well tolerated with adverse events comparable to those typically seen in the post-transplant population.

Source: Company Presentation

In addition to its HSCT-TMA lead program, Omeros is conducting ongoing Phase 3 clinical trials of Narsoplimab for immunoglobulin A nephropathy and atypical hemolytic uremic syndrome. However, due to COVID-19, enrollment has slowed while previously enrolled patients are continuing in the trials. Despite this, Omeros recently reaffirmed that they are targeting next year for the IgA nephropathy trial readout.

Source: Company Presentation

Looking ahead, Omeros believes that they will complete their BLA sometime this quarter. Given the Breakthrough Therapy designation, it's expected that the FDA will grant the BLA priority review. After FDA approval, the company will submit their European marketing authorization application. Commercial preparations are underway. The company is busy finalizing distribution, pricing, their launch strategy, hiring team members, and performing market research. Also, Omeros is assessing expansion of Narsoplimab into other endothelial injury syndromes, like diffuse alveolar hemorrhage and Graft-versus-host disease.

Source: Company Presentation

As of March 31st, 2020, Omeros had cash, cash equivalents and short-term investments of $53.9 million, compared to $60.7 million on December 31st, 2019. Research and development expenses for the first quarter were $28.9 million, compared to $26.2 million in Q1 of 2019. Selling, general and administrative expenses were $18 million in the quarter, compared to $14.6 million in the same quarter of 2019. Product sales came in at $23.5 million, compared to $21.7 million in Q1 of 2019. Record sales of $33.4 million were hit in Q4 of 2019, but they have since declined due to inventory utilization by ASCs and hospitals in anticipation of the COVID-19-related shutdown of elective surgical procedures. Overall, the company reported a net loss of $29 million for Q1 of 2020, compared to a net loss of $24.3 million for Q1 of 2019. The last time the company raised money was when they launched an equity offering in December of 2019.

Analysts are mixed on Omeros at the moment with three Buy ratings and two Holds over the past few months. The consensus price target on Wall Street is just north of $25.00, however. The latest rating comes from Wedbush on June 9th. The firm reiterated their hold rating and placed a $17 price target on the name. On May 12th, Needham & Company reiterated their hold rating. Lastly, on the same day as Needham & Company, HC Wainwright reiterated their buy rating and $34 price target. HC Wainwright has long had a price target in the $30's. In the analyst's valuation model, Omidria makes up roughly a third of the value and Narsoplimab makes up the other two thirds. Risks to the $34 price target, as outlined by the analyst at HC Wainwright, include: "(1) trial delays; (2) adverse clinical results; (3) inability to obtain approval for OMS721 and other candidates; (4) inability to achieve more OMIDRIA market traction; and (5) earlier-than-anticipated introduction of OMIDRIA generics in the U.S."

There are a couple of near term potential catalysts on the horizon. The first is completion of the company's rolling BLA on Narsoplimab for its first of hopefully many indications. The second is getting pass through status for Omidria past September 30th when it expires. Management sounded very confident this was going to happen by summer through a legislative effort that would garner Omidria the coveted status for an additional five years. The stock should get a boost from both of these events.

I think the company could then raise additional capital via a secondary offering to fund the rollout of Narsoplimab which I still believe has blockbuster potential. A good way to add exposure to Omeros is via a covered call strategy like the one outlined below.

Option Strategy

Here is how I have added exposure to OMER within my own personal accounts to make money even if the shares remain range bound. Using the February $15 call strikes fashion a Buy-Write order with a net debit in the $11.00 to $11.50 range (net stock price - option premium). This strategy provides approximately 20% downside risk (at the midpoint of the range) and just over 30% potential return in approximately seven months, again at the midpoint of the range. Liquidity in this name can vary significantly from day to day, so you may have to place your order more than once to fill.

The sad truth about humanity...is that people believe what they're told. Maybe not the first time, but by the hundredth time, the craziest of ideas just becomes a given." Neal Shusterman, UnWholly

Bret Jensen is the Founder of and authors articles for the Biotech Forum, Busted IPO Forum, and Insiders Forum

Live Chat on The Biotech Forum has been dominated by discussion of these type of buy-write opportunities over the past several months. To see what I and the other season biotech investors are targeting as trading ideas real-time, just join our community atThe Biotech Forum by clicking HERE.

Disclosure: I am/we are long OMER. I wrote this article myself, and it expresses my own opinions. I am not receiving compensation for it (other than from Seeking Alpha). I have no business relationship with any company whose stock is mentioned in this article.

See the original post:
The Play On Omeros - Seeking Alpha

2020 Memorial Tournament live stream, TV coverage, Round 3 tee times, watch Tiger Woods on Saturday – CBS Sports

The spotlight of the golf world this weekend is on an event akin to a major championship as the 2020 Memorial Tournament concludes over two days at Muirfield Village in Ohio. Golf fans had better buckle up because with Tiger Woods barely making it inside the cut, Bryson DeChambeau ejecting beyond the knee number, and a strong leaderboard that includes Tony Finau, Jon Rahm, Jordan Spieth and Jason Day all inside the top 10, anything can happen over the weekend.

A week after hosting the first Workday Charity Open on the same course, Murifield Village bared its teeth a bit on Thursday and Friday with scoring plummeting. Finau and Ryan Palmer sit at T1 after 36 holes, one shot up on Rahm. There's 12 other golfers within five shots of the leaders, too.

Keep on reading for how you can watch all of the action this weekend. Check out where we stand after Round 2, Woods' rough Friday and DeChambeau going full "Tin Cup" with a 10 on the 15th hole.All times Eastern; streaming start times approximated.

Round 3 starts:8:30 a.m. Featured groups and holes:8:40 a.m. - 6 p.m. --PGA Tour Live Early TV coverage: 12:30-3 p.m. on Golf Channel

Live TV coverage:3-6 p.m. on CBS Live simulcast:3-6 p.m. onCBSSports.comand theCBS Sports App Radio:1-6 p.m. onPGA Tour Radio

Round 4 starts:8:30 a.m. Featured groups and holes:8:40 a.m. - 7 p.m. --PGA Tour Live Early TV coverage:1-3:30 p.m. on Golf Channel

Live TV coverage:3:30-7 p.m. on CBS Live simulcast:3:30-7 p.m. onCBSSports.comand theCBS Sports App Radio:2-7 p.m. onPGA Tour Radio

Play along with this week's golf action andcompete to win $1,000each round with CBS Sports Golf Props. All you need to do is answer a few quick questions and you're in contention for the cash! Join any round for your chance to win. Terms apply.

7:30 a.m. -- Patrick Reed 7:40 a.m. -- Abraham Ancer, Xander Schauffele 7:50 a.m. -- Corey Conners, Stewart Cink 8:00 a.m. -- Tiger Woods, Brooks Koepka 8:10 a.m. -- Brendon Todd, Billy Horschel 8:20 a.m. -- Joel Dahmen, Marc Leishman 8:30 a.m. -- C.T. Pan, Kevin Streelman 8:40 a.m. -- Cameron Smith, Charles Howell III 8:50 a.m. -- Bubba Watson, Collin Morikawa 9:00 a.m. -- Sung Kang, Keegan Bradley 9:10 a.m. -- Mark Hubbard, Carlos Ortiz 9:20 a.m. -- Denny McCarthy 9:30 a.m. -- Adam Hadwin, Phil Mickelson 9:40 a.m. -- Tyler Duncan, Zach Johnson 9:50 a.m. -- Si Woo Kim, Bud Cauley 10:00 a.m. -- William McGirt, Vijay Singh 10:10 a.m. -- Sebastian Munoz, Keith Mitchell 10:20 a.m. -- Scott Piercy, Matthew Wolff 10:30 a.m. -- Xinjun Zhang, Ryan Moore 10:40 a.m. -- Louis Oosthuizen, Sepp Straka 10:50 a.m. -- Jason Dufner, Sergio Garcia 11:00 a.m. -- Erik van Rooyen, Lanto Griffin 11:10 a.m. -- Scottie Scheffler, Carl Pettersson 11:20 a.m. -- Harris English 11:30 a.m. -- Brendan Steele, Matt Kuchar 11:40 a.m. -- Kevin Na, Scott Harrington 11:50 a.m. -- Bo Hoag, Patrick Rodgers 12:00 p.m. -- Rory McIlroy, Matt Wallace 12:10 p.m. -- Justin Thomas, Jimmy Walker 12:20 p.m. -- Dylan Frittelli 12:30 p.m. -- Christiaan Bezuidenhout, Matthew Fitzpatrick 12:40 p.m. -- Lucas Glover, Patrick Cantlay 12:50 p.m. -- Jordan Spieth, Viktor Hovland 1:00 p.m. -- Danny Willett, Jim Furyk 1:10 p.m. -- Steve Stricker, Henrik Norlander 1:20 p.m. -- Jason Day, Mackenzie Hughes 1:30 p.m. -- Luke List, Chez Reavie 1:40 p.m. -- Jon Rahm, Gary Woodland 1:50 p.m. -- Ryan Palmer, Tony Finau

Visit link:
2020 Memorial Tournament live stream, TV coverage, Round 3 tee times, watch Tiger Woods on Saturday - CBS Sports

A synergistic role of convalescent plasma and mesenchymal stem cells in the treatment of severely ill COVID-19 patients: a clinical case report -…

This article was originally published here

Stem Cell Res Ther. 2020 Jul 16;11(1):291. doi: 10.1186/s13287-020-01802-8.

ABSTRACT

Acute respiratory distress syndrome virus-2 (SARS-CoV-2) responsible for coronavirus disease 2019 (COVID-19) infection, which causes global public health emergencies, has sped widely for more than 5 months and has the risk of long-term transmission. No effective treatment has been discovered to date. In the cases we report, the patient continued to deteriorate even after administration of antiviral drugs such as lopinavir/ritonavir, interferon-, and ribavirin, as well as intravenous injection of meropenem, methylprednisolone, and immunoglobulin. So, we infused the patient with convalescent plasma (CP), and the absolute lymphocyte count increased the next day and returned to normal on the fourth day. Followed by intravenous infusion of mesenchymal stem cells (MSCs), bilateral infiltrates were absorbed and the pulmonary function was significantly improved. We note that the intravenous infusion of CP and MSCs for the treatment of severe COVID-19 patients may have synergistic characteristics in inhibiting cytokine storm, promoting the repair of lung injury, and recovering pulmonary function. We hope to provide a reference for the research direction of COVID-19 clinical strategies.

PMID:32678017 | DOI:10.1186/s13287-020-01802-8

Original post:
A synergistic role of convalescent plasma and mesenchymal stem cells in the treatment of severely ill COVID-19 patients: a clinical case report -...