Categories
- Global News Feed
- Uncategorized
- Alabama Stem Cells
- Alaska Stem Cells
- Arkansas Stem Cells
- Arizona Stem Cells
- California Stem Cells
- Colorado Stem Cells
- Connecticut Stem Cells
- Delaware Stem Cells
- Florida Stem Cells
- Georgia Stem Cells
- Hawaii Stem Cells
- Idaho Stem Cells
- Illinois Stem Cells
- Indiana Stem Cells
- Iowa Stem Cells
- Kansas Stem Cells
- Kentucky Stem Cells
- Louisiana Stem Cells
- Maine Stem Cells
- Maryland Stem Cells
- Massachusetts Stem Cells
- Michigan Stem Cells
- Minnesota Stem Cells
- Mississippi Stem Cells
- Missouri Stem Cells
- Montana Stem Cells
- Nebraska Stem Cells
- New Hampshire Stem Cells
- New Jersey Stem Cells
- New Mexico Stem Cells
- New York Stem Cells
- Nevada Stem Cells
- North Carolina Stem Cells
- North Dakota Stem Cells
- Oklahoma Stem Cells
- Ohio Stem Cells
- Oregon Stem Cells
- Pennsylvania Stem Cells
- Rhode Island Stem Cells
- South Carolina Stem Cells
- South Dakota Stem Cells
- Tennessee Stem Cells
- Texas Stem Cells
- Utah Stem Cells
- Vermont Stem Cells
- Virginia Stem Cells
- Washington Stem Cells
- West Virginia Stem Cells
- Wisconsin Stem Cells
- Wyoming Stem Cells
- Biotechnology
- Cell Medicine
- Cell Therapy
- Diabetes
- Epigenetics
- Gene therapy
- Genetics
- Genetic Engineering
- Genetic medicine
- HCG Diet
- Hormone Replacement Therapy
- Human Genetics
- Integrative Medicine
- Molecular Genetics
- Molecular Medicine
- Nano medicine
- Preventative Medicine
- Regenerative Medicine
- Stem Cells
- Stell Cell Genetics
- Stem Cell Research
- Stem Cell Treatments
- Stem Cell Therapy
- Stem Cell Videos
- Testosterone Replacement Therapy
- Testosterone Shots
- Transhumanism
- Transhumanist
Archives
Recommended Sites
Monthly Archives: November 2020
UB researcher narrows time window for administering specific treatment to infants with Krabbe disease – UB Now: News and views for UB faculty and…
Posted: November 13, 2020 at 4:56 am
A team of UB researchers has published a paper in Nature Communications that is helping to define the best time to give a specific treatment to infants born with Krabbe disease (KD). This treatment has been found to prolong life for these infants for as long as a few years.
The paper was published online in Nature Communication Oct. 23.
Daesung Shin, assistant professor in the Department of Biotechnical and Clinical Laboratory Sciences and the Neuroscience Program, both in the Jacobs School of Medicine and Biomedical Sciences at UB, is the lead investigator. He also conducts research at UBs Hunter James Kelly Research Institute.
KD is an inherited disorder that destroys myelin, the protective coating of nerve cells in the brain and throughout the nervous system. In most cases, signs and symptoms of Krabbe disease develop in babies before 6 months of age, and the disease usually results in death by age 2. When it develops in older children and adults, the course of the disease can vary greatly.
The progressive neurologic disorder is caused by a deficiency of galactosylceramidase (GALC). GALC is an enzyme that breaks down galactosylceramide, an important component of myelin, which ensures the rapid transmission of nerve impulses.
Although there is no cure for KD, hematopoietic stem cell therapy (HSCT), a therapy that makes blood cells, reduces neurologic deterioration and improves developmental advances. These benefits are dependent on the severity of the disease at the time the stem cells are transplanted, and are only beneficial if delivered at a clinically defined pre-symptomatic time point before symptoms appear.
Even though it is widely accepted that early treatment is essential for the most positive outcome, the precise therapeutic window for treatment and what happens during this early time have never been elucidated, Shin says.
To address that issue, his team used mutations to create a novel mouse model of KD.
We engineered an inducible knockout mouse for the GALC gene deletion in specific cells at specific times, which provided us with the opportunity to directly ask when and where GALC enzyme is required for brain development, Shin explains.
We were particularly interested in the role of early developmental GALC function, he says. Our study not only revealed a key developmental process that requires GALC in the perinatal period, but also demonstrated that temporal GALC expression is likely a major contributor to brainstem development.
The researchers found that by increasing GALC levels at or before this newly defined perinatal period they could improve the effectiveness of therapeutic interventions for KD.
For the first time, our work showed the mechanistic evidence to explain why treatment must occur so early, with the defined critical postnatal period at days 4-6 in mice, and demonstrated that temporal GALC expression during this time is a major contributor to brainstem development, Shin says. Augmenting GALC levels at or prior to this newly defined period would likely improve the efficacy of therapeutic interventions for Krabbe patients.
While the time scale between mice and humans is considerably different, the sequence of key events in brain maturation between the two is consistent, he notes. It was estimated that the mouse nervous system at postnatal days 4-6 corresponds to a gestational age of 32 weeks in humans. Therefore, we anticipate that if our result is correct, then in utero treatments at, or prior to, 32 weeks should have better outcomes than conventional postnatal treatment for Krabbe babies.
Shin says his team will further identify which cell type needs to be targeted with therapy.
This work will directly impact the design of novel treatment options for KD patients, he says, noting that KD studies are at the basis of research on other, more common neurodegenerative diseases, such as multiple sclerosis and Parkinsons disease. Therefore, the teams work will have implications beyond KD.
Co-authors on the research were Nadav I. Weinstock, MD-PhD student, and Conlan Kreher, former masters student, both of the HJKRI and the Department of Biochemistry in the Jacobs School; Jacob Favret, research technician in the Department of Biotechnical and Clinical Laboratory Sciences; Lawrence Wrabetz and M. Laura Feltri, both co-directors of the HJKRI and members of the departments of Biochemistry and Neurology, as well as the Neuroscience Program.
Duc Nguyen and Ernesto R. Bongarzone of the Department of Anatomy and Cell Biology in the College of Medicine at the University of Illinois at Chicago also participated in the research.
The project was initiated with the support from Empire State development fund for HJKRI, and further developed and finalized by the R01, R56 and R03 grants from National Institutes for Health-National Institute for Neurological Disorders and Stroke awarded to Shin.
See the article here:
UB researcher narrows time window for administering specific treatment to infants with Krabbe disease - UB Now: News and views for UB faculty and...
Posted in Stem Cell Treatments
Comments Off on UB researcher narrows time window for administering specific treatment to infants with Krabbe disease – UB Now: News and views for UB faculty and…
Addenbrooke’s becomes first regional centre to offer CAR-T cell treatment for cancer patients – Cambridge Network
Posted: November 13, 2020 at 4:56 am
The decision to offer the therapy puts Cambridge University Hospitals in the premier league of world class cancer centres.
It also supports progress of the new Cambridge Cancer Research Hospital on the Addenbrookes site, which the Government confirmed in October is included in the second wave of the Hospital Infrastructure Programme (HIP2).
To begin with, the pioneering treatment will be offered to patients with aggressive B-cell lymphomas and acute lymphoblastic leukaemia (ALL) who have relapsed or not responded well to chemotherapy or stem cell treatment.
It is also likely to be offered to cancer patients aged over 70 who are considered to be too high risk to have stem cell transplants.
CAR-T cell therapy can have extremely positive results, but because it can cause unpleasant side effects, it tends to be used when there are few other treatment options available.
CAR-T cell therapy works by re-engineering the patients own immune cells.
Our immune cells, which are called T-cells, circulate around the bloodstream seeking out and destroying any alien intruders, such as infections.
But because cancer cells evolve from our own cells, sometimes our T-cells do not identify them as intruders, and leave them alone.
The new CAR-T cell therapy works by harvesting a patients own T-cells. These are sent to a specialist lab, where they are reprogrammed to express a molecule on their surface, called a Chimeric Antigen Receptor, or CAR, that targets them to the cancer. The reprogrammed cells are grown in a huge numbers over a few weeks and then infused back into the patients body, where they seek out and destroy the cancer. Its a bit like giving immune cells a sat nav.
Until now patients in the east of England needing CAR-T cell therapy have had to make frequent trips to London in preparation for the treatment, which can take another month to administer.
One such patient was Steve Johnson from Bourne who underwent CAR-T cell therapy for relapsed leukaemia at the University College Hospital in London as part of a clinical trial.
Steve explained: "Having the treatment is not pleasant I had a number of fevers and temperature spikes for two weeks after the CAR-T cells were put back in, but I have absolutely no doubt this treatment saved my life and without it I would not be here today."
Steve added: I was lucky for me the trial came at the right time. Having the option to explore and provide revolutionary treatments at places like Addenbrookes and the soon to be built Cambridge Cancer Research Hospital, is vital if we are going to rewrite the story of this devastating illness.
The first patients to be approved for CAR-T cell treatment at Addenbrookes will start having their cells harvested from this week.
Ben Uttenthal, consultant haematologist specialising in CAR-T cell therapy at Addenbrookes, said: This is an exciting new way of treating patients that attacks cancer in a different way from previously available medicines.
"Being able to offer CAR-T cell therapy in our armoury against cancer puts us in the premier league of cancer centres.
It is also a testament to the expertise available across many different specialties in Addenbrookes. Through offering treatments like this, now and in the new Cambridge Cancer Research Hospital, we will be able to benefit cancer patients locally, regionally and nationally.
Initially, the hospital expects to be able to offer CAR-T cell therapy to around 40 patients a year. In the future, treatment is likely to be expanded to include patients with other cancers.
View original post here:
Addenbrooke's becomes first regional centre to offer CAR-T cell treatment for cancer patients - Cambridge Network
Posted in Stem Cell Treatments
Comments Off on Addenbrooke’s becomes first regional centre to offer CAR-T cell treatment for cancer patients – Cambridge Network
"Revolutionary" cancer treatment to be offered at Addenbrooke’s Hospital Cambridge – In Your Area
Posted: November 13, 2020 at 4:56 am
By Alya Zayed
Pictured above: Addenbrookes patient Steve Johnson who received the new treatment. Photo credit: Addenbrooke's Hospital.
A revolutionary cancer treatment that could work on patients not responding to chemotherapy will be offered by Addenbrookes Hospital.
The hospital will be the first in East Anglia to offer CAR-T cell treatment, which puts it in the premier league of cancer centres, said one doctor from Cambridge University Hospitals NHS Foundation Trust, which runs the hospital.
The pioneering treatment will be offered to patients with some cancers who have relapsed or not responded well to chemotherapy or stem cell treatment - namely, aggressive B-cell lymphomas and acute lymphoblastic leukaemia (ALL).
It is also likely to be offered to cancer patients aged over 70 who are considered to be too high risk to have stem cell transplants.
The new therapy can have extremely positive results, but is used as a last resort because of its unpleasant side effects, such as high fevers and low blood pressure.
CAR-T cell therapy works by re-engineering the patients own immune cells.
Immune cells, which are called T-cells, circulate around the bloodstream seeking out and destroying any intruders, such as infections.
But because cancer cells evolve from our own cells, sometimes T-cells do not identify them as intruders, and leave them alone.
The new CAR-T cell therapy works by harvesting a patients own T-cells. These are sent to a specialist lab, where they are reprogrammed to express a molecule on their surface, called a Chimeric Antigen Receptor, or CAR, that targets them to the cancer.
The reprogrammed cells are grown in huge numbers over a few weeks and then infused back into the patients body, where they seek out and destroy the cancer.
It can be a bit like giving immune cells a Sat-Nav, explained the hospital.
The first patients to be approved for CAR-T cell treatment at Addenbrookes will start having their cells harvested from this week.
Initially, the hospital expects to be able to offer CAR-T cell therapy to around 40 patients a year.
In the future, treatment is likely to be expanded to include patients with other cancers.
Ben Uttenthal, consultant haematologist specialising in CAR-T cell therapy at Addenbrookes, said: This is an exciting new way of treating patients that attacks cancer in a different way from previously available medicines.
Being able to offer CAR-T cell therapy in our armoury against cancer puts us in the premier league of cancer centres.
It is also a testament to the expertise available across many different specialties in Addenbrookes.
Through offering treatments like this, now and in the new Cambridge Cancer Research Hospital, we will be able to benefit cancer patients locally, regionally and nationally.
Until now, patients in East Anglia have had to travel to London for the treatment.
Addenbrookes patient Steve Johnson, from Bourn, south Cambridgeshire, is one of many who went to University College Hospital, London, to take part in a clinical trial.
Steve, whose leukemia had relapsed, said: Having the treatment is not pleasant I had a number of fevers and temperature spikes for two weeks after the CAR-T cells were put back in, but I have absolutely no doubt this treatment saved my life and without it I would not be here today.
I was lucky for me the trial came at the right time. Having the option to explore and provide revolutionary treatments at places like Addenbrookes and the soon to be built Cambridge Cancer Research Hospital, is vital if we are going to rewrite the story of this devastating illness.
The new Cambridge Cancer Research Hospital on the Addenbrookes site was confirmed by the government in October as one of the new hospitals to be built.
See the latest news, information, conversations and much more, all tailored to your neighbourhood, in your InYourArea live feed here.
More:
"Revolutionary" cancer treatment to be offered at Addenbrooke's Hospital Cambridge - In Your Area
Posted in Stem Cell Treatments
Comments Off on "Revolutionary" cancer treatment to be offered at Addenbrooke’s Hospital Cambridge – In Your Area
Orchard Therapeutics Unveils Details on New HSC Gene Therapy Research Programs as Part of R&D Investor Event Tomorrow at 9:00 a.m. ET – GlobeNewswire
Posted: November 13, 2020 at 4:56 am
First look at preclinical data in frontotemporal dementia with progranulin mutations (GRN-FTD) and new amyotrophic lateral sclerosis (ALS) program
NOD2 mutation revealed as Crohns disease (CD) genetic target, associated with 7-10% of all CD cases in the U.S. and Europe
Deep dive on transduction enhancers and stable cell line technology innovations that support manufacturing for larger indications
BOSTON and LONDON, Nov. 12, 2020 (GLOBE NEWSWIRE) -- Orchard Therapeutics (Nasdaq: ORTX), a global gene therapy leader, today previewed details on its investigational hematopoietic stem cell (HSC) gene therapy research programs in GRN-FTD and NOD2-CD in advance of an upcoming virtual R&D investor event. The company also disclosed a new research program in ALS. A live webcast of the presentation will be available in the Investors & Media section of the companys website at http://www.orchard-tx.com starting Friday, November 13, 2020 at 9:00 a.m. ET.
We are excited to draw back the curtain at tomorrows event on our work in larger indications that form an important part of Orchards evolution as a company, including a new program in ALS, in addition to our work in genetic subsets of FTD and Crohns disease, said Bobby Gaspar, M.D., Ph.D., chief executive officer, Orchard Therapeutics. These research programs have been established using a scientific approach that has resulted in more than 160 patients being treated across multiple rare diseases and a recent positive CHMP opinion in the EU for Libmeldy. We believe that HSC gene therapy has the power to transform lives, and we are excited about the possibilities for Orchard and patients with its expanded application.
OTL-204 for GRN-FTD and new ALS research program
The GRN-FTD and ALS programs are based on the same HSC gene therapy approach that has been clinically validated with Libmeldy (OTL-200), Orchards program for metachromatic leukodystrophy, and is under clinical evaluation in the OTL-203 and OTL-201 programs for mucopolysaccharidosis type I and mucopolysaccharidosis type IIIA, respectively. Development work in GRN-FTD and ALS will be undertaken as part of a collaboration with Boston Childrens Hospital (BCH), the University of Padua (UNIPD) and Prof. Alessandra Biffi, chair of the Pediatric Hematology, Oncology and Stem Cell Transplant Division at UNIPD and co-director of the Gene Therapy Program at BCH.
Prof. Biffi commented, The ability of HSC gene therapy to restore healthy microglia function supports the use of this technology for the development of treatments for a variety of diseases with central nervous system involvement. In GRN-FTD, initial in vitro data shows progranulin expression and secretion in culture and uptake indicative of cross-correction. My previous work at BCH researching ALS supports the novel approach of treating this severe neurodegenerative condition by targeting the NOX2 pathway.
OTL-104 for NOD2-CD
Orchards preclinical program in CD targets mutations in the nucleotide-binding oligomerization domain-containing protein 2 (NOD2) gene, which plays a role in immune cell response to bacterial peptides in the gastrointestinal (GI) tract. The companys proposed approach leverages this link, using gene modified HSC-derived cells (monocytes) to replace GI resident macrophages, thus potentially correcting the inflammation and colitis associated with NOD2-CD.
Manufacturing Innovations to Support Work in Larger IndicationsTransduction enhancers (TEs) and stable cell line technology (SCLT)
Orchard has completed a thorough TE screening process and identified and validated several novel TE compounds, which in combination, facilitate lentiviral vector entry into HSCs and have shown a greater than 50% reduction in vector requirements. The enhancers mode of action is expected to be effective in each of Orchards HSC gene therapy programs. An evaluation of enhancer-treated HSC engraftment potential in mice is currently underway.
The company has worked extensively with SCLT, including the technology licensed from GSK for certain programs, to both develop processes to efficiently create SCLs for new vectors and scale up the production of SCLs to clinical grade. Results have delivered consistent levels of high-titer lentiviral production comparable to those seen using conventional methods. Selection of single high-titer clones for new vectors using this method has been achieved within three months. Work at Orchard is ongoing to develop upstream and downstream processes to further improve productivity and scalability.
We have a clear roadmap for Orchards future that prioritizes strategic growth and draws on the many synergies across our scientific, manufacturing and emerging commercial platforms, said Frank Thomas, president and chief operating officer. Over the next 12 months we have an array of exciting commercial, regulatory and clinical milestones that will continue to showcase the breadth and depth of our advanced HSC gene therapy portfolio.
Webcast Information
A live webcast of the presentation New Horizons in Gene Therapy will be available under Events in the Investors & Media section of the companys website at http://www.orchard-tx.com. A replay of the webcast will be archived on the Orchard website following the presentation.
About Orchards Research Collaborations
In connection with its previously disclosed collaboration with Prof. Alessandra Biffi, Orchard has signed agreements with Boston Childrens Hospital and the University of Padua to develop and exclusively license new ex vivo HSC gene therapy programs, patents and technologies for the treatment of neurodegenerative disorders. As part of the collaboration, Orchard has initiated sponsored research agreements and obtained exclusive options to license multiple new preclinical programs, including frontotemporal dementia with progranulin mutations (GRN-FTD), amyotrophic lateral sclerosis (ALS) and other rare and less rare indications.Orchard continues to support Professor Biffis labs in the development of new proprietary technology focused on enhancing the application of gene-modified HSC therapy for CNS disorders.
About Orchard
Orchard Therapeutics is a global gene therapy leader dedicated to transforming the lives of people affected by rare diseases through the development of innovative, potentially curative gene therapies. Our ex vivo autologous gene therapy approach harnesses the power of genetically modified blood stem cells and seeks to correct the underlying cause of disease in a single administration. In 2018, Orchard acquired GSKs rare disease gene therapy portfolio, which originated from a pioneering collaboration between GSK and theSan Raffaele Telethon Institute for Gene Therapy in Milan, Italy. Orchard now has one of the deepest and most advanced gene therapy product candidate pipelines in the industry spanning multiple therapeutic areas where the disease burden on children, families and caregivers is immense and current treatment options are limited or do not exist.
Orchard has its global headquarters in London and U.S. headquarters in Boston. For more information, please visit http://www.orchard-tx.com, and follow us on Twitter and LinkedIn.
Availability of Other Information About Orchard
Investors and others should note that Orchard communicates with its investors and the public using the company website (www.orchard-tx.com), the investor relations website (ir.orchard-tx.com), and on social media (Twitter and LinkedIn), including but not limited to investor presentations and investor fact sheets, U.S. Securities and Exchange Commission filings, press releases, public conference calls and webcasts. The information that Orchard posts on these channels and websites could be deemed to be material information. As a result, Orchard encourages investors, the media, and others interested in Orchard to review the information that is posted on these channels, including the investor relations website, on a regular basis. This list of channels may be updated from time to time on Orchards investor relations website and may include additional social media channels. The contents of Orchards website or these channels, or any other website that may be accessed from its website or these channels, shall not be deemed incorporated by reference in any filing under the Securities Act of 1933.
Forward-Looking Statements
This press release contains certain forward-looking statements about Orchards strategy, future plans and prospects, which are made pursuant to the safe harbor provisions of the Private Securities Litigation Reform Act of 1995. Such forward-looking statements may be identified by words such as anticipates, believes, expects, plans, intends, projects, and future or similar expressions that are intended to identify forward-looking statements.Forward-looking statements include express or implied statements relating to, among other things, Orchards business strategy and goals, including with respect to its manufacturing strategy, expected future milestones, and its plans and expectations for the development of its product candidates, including the product candidates referred to in this release, and the therapeutic and commercial potential of its product candidates.These statements are neither promises nor guarantees and are subject to a variety of risks and uncertainties, many of which are beyond Orchards control, which could cause actual results to differ materially from those contemplated in these forward-looking statements. In particular, these risks and uncertainties include, without limitation: the risk that any one or more of Orchards product candidates, including the product candidates referred to in this release, will not be approved, successfully developed or commercialized; the risk that prior results, such as signals of safety, activity or durability of effect, observed from preclinical studies or clinical trials of Orchards product candidates will not be repeated or continue in ongoing or future studies or trials involving its product candidates; the risk that the market opportunity for its product candidates may be lower than estimated; and the severity of the impact of the COVID-19 pandemic on Orchards business, including on preclinical and clinical development, its supply chain and commercial programs. Given these uncertainties, the reader is advised not to place any undue reliance on such forward-looking statements.
Other risks and uncertainties faced by Orchard include those identified under the heading Risk Factors in Orchards quarterly report on Form 10-Q for the quarter ended September 30, 2020, as filed with the U.S. Securities and Exchange Commission (SEC), as well as subsequent filings and reports filed with the SEC. The forward-looking statements contained in this press release reflect Orchards views as of the date hereof, and Orchard does not assume and specifically disclaims any obligation to publicly update or revise any forward-looking statements, whether as a result of new information, future events or otherwise, except as may be required by law.
Contacts
InvestorsRenee LeckDirector, Investor Relations+1 862-242-0764Renee.Leck@orchard-tx.com
MediaChristine HarrisonVice President, Corporate Affairs+1 202-415-0137media@orchard-tx.com
1Knopman DS, Roberts RO. J Mol Neurosci. 2011, Onyike CU, Diehl-Schmid J. Int Rev Psychiatry. 2013 and Riedl L, et al Neuropsychiatr Dis Treat. 20142 Centers for Disease Control and Prevention; European Crohns and Colitis Organisation (ECCO); Ashton, James J et al.Clin Transl Gastroenterol. 2020 Feb
Continue reading here:
Orchard Therapeutics Unveils Details on New HSC Gene Therapy Research Programs as Part of R&D Investor Event Tomorrow at 9:00 a.m. ET - GlobeNewswire
Posted in Stem Cell Treatments
Comments Off on Orchard Therapeutics Unveils Details on New HSC Gene Therapy Research Programs as Part of R&D Investor Event Tomorrow at 9:00 a.m. ET – GlobeNewswire
CAR T-Cell Therapy for Multiple Myeloma: What Community Oncologists Need to Know – OncLive
Posted: November 13, 2020 at 4:56 am
Multiple myeloma is a plasma cell malignancy that accounts for approximately 1% of new cancer diagnoses per year. The development of a combination approach of mainstay therapies including immunomodulatory drugs (IMiDs), proteasome inhibitors (PIs), and antiCD-38 antibodies has greatly improved up-front remission rates and response duration and prolonged overall survival in the past decade. However, for patients who have been heavily exposed to or are triple or penta-refractory to these classes of therapy, survival is under 1 year.1 Therefore, the emergence of cellular-based therapies represents a major opportunity to improve outcomes in the heavily pretreated and refractory myeloma population.
Chimeric antigen receptor (CAR) T-cell therapies approved by the FDA for several relapsed and refractory B-cell lymphomas are presently under investigation in a variety of treatment line settings for multiple myeloma. As more patients in the community become eligible for CAR T-cell therapies, community oncologists will need to be increasingly familiar about the various products, including their immediate and longer-term risks. In addition, it is key to understand the optimal time for referring these patients to an academic institution, as well as how to manage the requisite post CAR T-cell therapy in the community setting.
The major CAR T-cell therapies in trials for multiple myeloma are directed toward B-cell maturation antigen (BCMA), a surface antigen expressed on B cells starting from their period of development in the germinal centers onward and persisting through the time of plasma cell differentiation. BCMA is minimally expressed in normal human tissues but is heavily expressed on myeloma plasma cells. This allows for safe and intentional targeting of neoplastic myeloma cells using CAR T-cell technology.
The first FDA-approved commercial CAR T-cell therapy will likely arrive in the first quarter of 2021. Patients enrolled on the initial BCMA CAR T-cell trials were required to have had 3 or more prior lines of therapy with exposure to a PI, an IMiD, and an anti-CD38 monoclonal antibody.
The earliest trial was the KarMMa-1 study (NCT03361748) of idecabtagene vicleucel (ide-cel, previously bb2121), which demonstrated an overall response rate (ORR) of 73% and a median progression-free survival (PFS) of 8.8 months in all dose levels. Additionally, a median PFS of 12.1 months was observed in the highest dose level of CAR T cells (450 106).2,3 Patients on this trial (n = 128) had a median of 6 prior lines of therapy, and 84% were triple refractory.
A concurrent trial, CARTITUDE-1 (NCT03548207), studying JNJ-4528 (ciltacabtagene autoleucel in China), also targeting BCMA, demonstrated an unprecedented 100% ORR in patients, with 86% achieving stringent complete response (sCR), the deepest response level by International Myeloma Working Group criteria.4 In the most recent evaluation of PFS, 86% of patients on CARTITUDE-1 had not progressed in disease at a median follow-up time of 9 months.5 New data for JNJ-4528 and the other CAR T-cell therapies being investigated will be made available during the 2020 American Society of Hematology Annual Meeting and Exposition in December and should further reinforce the potential for CAR T-cell therapy to deliver deep and durable responses to patients with multiple myeloma who are heavily pretreated.
What Type of Patients Might Benefit the Most From CAR T?
As in the case of BCMA, antigens for CAR T cells are chosen for their unique expression in tumor tissue and low expression in normal, healthy tissue. The complications that occur in patients receiving CAR T-cell therapy will be largely specific to the targeted antigen. Patients must undergo a multistep process to safely and effectively undergo CAR T-cell treatment and should know up front about the process involved.
The initial step involves leukapheresis, or collecting the patients T cells. Once this is completed, the patient must wait 3 to 5 weeks for typical manufacturing of their CAR T cells at a production facility. If required, patients typically can undergo bridging chemotherapy during this waiting period to maintain their disease, particularly if myeloma progression is accelerating. Once the patients CAR T cells are produced, the patient will undergo lymphodepletion (typically with fludarabine/cyclophosphamide) for 3 days before being infused with prepared CAR T cells. Patients must have sufficient health reserve to withstand the potential adverse effects (AEs) associated with CAR T-cell therapy, including cytokine release syndrome (CRS) and immune effector cellassociated neurotoxicity syndrome (ICANS). Therefore, early referral to an academic center is key to getting these patients on therapy at the optimal time.
CRS is the most common acute AE that occurs in patients treated with CAR T-cell therapy. It occurred in nearly 9 out of 10 patients in the KarMMa-1 and CARTITUDE-1 trials, though severe CRS occurred only in 5% to 7% of patients.3,5 CRS is the phenotypic presentation of a supraphysiologic production of cytokines prompted by immune cell activation, including T cells, macrophages, and monocytes (Table 16 ). Preclinical models have shown that macrophages amplify CRS severity and are the source of several of the culprit cytokines of CRS, including interleukin (IL) 6, IL-1, and IFN-. CRS onset occurs variably depending on the CAR construct used.
Table 1. ASTCT Criteria for CRS6
For example, CRS occurred at median 1 day after infusion of ide-cel, whereas CRS occurred at median 7 days after infusion of JNJ-4528. Our primary treatment for CRS is tocilizumab (Actemra), a monoclonal antibody directed against IL-6; its use does not diminish efficacy of CAR T cells. Subsequent agents such as the IL-1 receptor antagonist anakinra (Kineret), antiIL-5 monoclonal antibody siltuximab (Sylvant), and corticosteroids may be employed for severe cases of CRS.
The second notable AE associated with CAR T-cell therapy is ICANS, which is defined by the American Society for Transplantation and Cellular Therapy as a disorder characterized by a pathological process involving the central nervous system.Symptoms or signs can be progressive and may include aphasia, altered level of consciousness, impairment of cognitive skills, motor weakness, seizures, and cerebral edema.6 A primarily encephalopathic picture appears when patients develop ICANS, which typically occurrs later than CRS. The primary treatment for ICANS involves corticosteroids such as dexamethasone, initially every 6 hours with a quick taper as symptoms start resolving.
Signs and symptoms of neurotoxicity should be screened at least daily during the acute CAR T-cell treatment period and can be assessed with validated tools such as those from CAR T-cell therapy-associated TOXicity (CARTOX) Working Group or the Immune-Effector Cell-Associated Encephalopathy (ICE) tool.6 Severe neurotoxicity occurred at only a 3% rate in KarMMa-1 and CARTITUDE-1 and can be treated readily if caught early with close monitoring.
Other common AEs such as cytopenia and infection may occur during and after the period of CAR T-cell infusion and expansion. Anemia, thrombocytopenia, or neutropenia may occur for months after the initial treatment period, and clinicians should utilize supportive transfusions and/or granulocyte colonystimulating factor and thrombopoietin mimetics as indicated.
Cytomegalovirus quantitative polymerase chain reaction should be checked monthly. If the patient is persistently neutropenic, opportunistic infections should be considered. Intravenous immunoglobulin should be administered before initial lymphodepletion, but also every month for 6 months after CAR T-cell infusion and during winter months if the patient has a history of recurrent infections. In addition, patients should be vaccinated at 1-year post CAR T-cell therapy; a recommended vaccine schedule is included in Table 2.
Table 2. Recommended Vaccine Schedules After CAR T-Cell Therapy
The multiple myeloma treatment landscape is rapidly evolving, with many exciting new therapies on the forefront for relapsed and refractory multiple myeloma. CAR T-cell therapy is a 1-time treatment that allows patients to achieve deep and durable remissions without the need for the usual continuous cancer-directed treatments. The aforementioned trials and therapies have paved the way for subsequent iterations of cellular therapies. These include nonBCMA-targeted CAR T cells; next-generation CAR T cells with improved costimulatory domains for improved safety, efficacy, or faster production; and allogeneic CAR T cells derived from pluripotent stem cells available off the shelf to patients. In the coming year, as commercially available myeloma CAR T-cell therapies are approved, it will be even more important for community oncologists to better understand these therapies so they can offer them to their patients.
References
Read the original:
CAR T-Cell Therapy for Multiple Myeloma: What Community Oncologists Need to Know - OncLive
Posted in Stem Cell Treatments
Comments Off on CAR T-Cell Therapy for Multiple Myeloma: What Community Oncologists Need to Know – OncLive
Leading Human Immunology and Infectious Disease Experts to Join UM School of Medicines Institute of Human Virology – Newswise
Posted: November 13, 2020 at 4:56 am
Newswise Baltimore, MD, November 12, 2020 Robert C. Gallo, MD, the Homer & Martha Gudelsky Distinguished Professor in Medicine at the University of Maryland School of Medicine (UMSOM) and Co-Founder & Director of the UMSOMs Institute of Human Virology (IHV), announced today that a team of leading scientists in human immunology, virology and stem cell biology, led by Lishan Su, PhD joined IHV on October 1 with academic appointments in the UMSOM Department of Pharmacology. As part of the Maryland E-Nnovation Initiative Fund (MEIF) to recruit top research faculty and a donation to IHV from the Charles Gordon Estate, Dr. Su has been named the Charles Gordon Smith Endowed Professor for HIV Research. Dr. Su will also head IHVs Division of Virology, Pathogenesis and Cancer.
The team will include a 12-person Laboratory of Viral Pathogenesis and Immunotherapy with two faculty appointments as well as major public and private sector research funding.
Dr. Gallo made the announcement in conjunction with University of Maryland School of Medicine Dean E. Albert Reece, MD, PhD, MBA and Margaret M. McCarthy PhD, James & Carolyn Frenkil Deans Professor, Chair of the Department of Pharmacology.
Dr. Su is one of the most successful active basic researchers in America, said Dr. Gallo, who is also Co-Founder and Chairman of the International Scientific Leadership Board of the Global Virus Network. His research is groundbreaking, and we are so pleased to have him join IHV and lead our Division of Infectious Agents and Cancer, which under his sound leadership, will flourish.
Dr. McCarthy added:Dr. Sus continuing ground-breaking work in HIV and Hepatitis B will be a huge asset to the Department of Pharmacology. I look forward to working with him on advances that could open the door to new therapeutics.
Dr. Su was a faculty member in the Lineberger Comprehensive Cancer Center and Professor in the Department of Microbiology & Immunology at University of North Carolina-Chapel Hill since 1996. He received his BS degree in Microbiology from Shandong University, his PhD degree in Virology from Harvard University, and did his post-doctoral training in Stem Cell Biology & Immunology at Stanford University. He worked as a senior research scientist at SyStemix/Sandoz (Novartis), focusing on HIV-1 pathogenesis and stem cell-based gene therapy in humanized mice and in patients.
I am excited to continue and expand my research programs at the Institute of Human Virology (IHV), said Dr. Su. I have long been impressed by the Baltimore-DC area's research centers with great basic and clinical research programs. IHV, co-founded and directed by Dr. Robert Gallo, is one of the first research institutes in the U.S. to integrate basic science, population studies and clinical trials to understanding and treating human virus-induced diseases. The Department of Pharmacology, headed by Dr. Margaret McCarthy, in the University of Maryland School of Medicine, has been outstanding in developing novel therapeutics including breast cancer drugs. I look forward to working with my new colleagues at IHV and the Department of Pharmacology, and across the University of Maryland School of Medicine, to expand and translate my research programs to treating human inflammatory diseases including virus infection and cancer.
Dr. Su has extensive research experience in human immunology, virology and stem cell biology. Dr. Su made important contributions to several areas of human immunology and infectious diseases, particularly in studying human immuno-pathology of chronic virus infections. His lab at UNC-Chapel Hill published important findings in identifying novel virological and immunological mechanisms of HIV-1 pathogenesis. Furthermore, his lab established humanized mouse models with both human immune and human liver cells that support HCV or HBV infection, human immune responses and human liver fibrosis. In recent years, Dr. Sus group discovered, and focused on, the pDC-interferon axis in the immuno-pathogenesis and therapy of chronic HIV & HBV infections. The group also started investigation of the pDC-IFN axis in tumor microenvironments and in cancer immune therapy.
Im so pleased to welcome Dr. Su to our faculty. His work advances the mission of the School of Medicine, which is to provide important new knowledge in the area of immunology and chronic disease to discover new approaches for treatments, said Dean Reece, who is also University Executive Vice President for Medical Affairs and the John Z. and Akiko K. Bowers Distinguished Professor. Dr. Sus stellar research capabilities will provide vital opportunities for collaboration across our Institutes and Departments.
About the Institute of Human Virology
Formed in 1996 as a partnership between the State of Maryland, the City of Baltimore, the University System of Maryland and the University of Maryland Medical System, IHV is an institute of the University of Maryland School of Medicine and is home to some of the most globally-recognized and world-renowned experts in all of virology. The IHV combines the disciplines of basic research, epidemiology and clinical research in a concerted effort to speed the discovery of diagnostics and therapeutics for a wide variety of chronic and deadly viral and immune disorders - most notably, HIV the virus that causes AIDS. For more information,www.ihv.organd follow us on Twitter @IHVmaryland.
About the University of Maryland School of Medicine
The University of Maryland School of Medicine was chartered in 1807 and is the first public medical school in the United States and continues today as an innovative leader in accelerating innovation and discovery in medicine. The School of Medicine is the founding school of the University of Maryland and is an integral part of the 11-campus University System of Maryland. Located on the University of Marylands Baltimore campus, the School of Medicine works closely with the University of Maryland Medical Center to provide a research-intensive, academic and clinically based education. With 43 academic departments, centers and institutes and a faculty of more than 3,000 physicians and research scientists plus more than $400 million in extramural funding, the School is regarded as one of the leading biomedical research institutions in the U.S. with top-tier faculty and programs in cancer, brain science, surgery and transplantation, trauma and emergency medicine, vaccine development and human genomics, among other centers of excellence. The School is not only concerned with the health of the citizens of Maryland and the nation, but also has a global vision, with research and treatment facilities in more than 30 countries around the world. For more information, visitwww.medschool.umaryland.edu.
Read more here:
Leading Human Immunology and Infectious Disease Experts to Join UM School of Medicines Institute of Human Virology - Newswise
Posted in Stem Cell Treatments
Comments Off on Leading Human Immunology and Infectious Disease Experts to Join UM School of Medicines Institute of Human Virology – Newswise
Angiocrine Bioscience Announces FDA Regenerative Medicine Advanced Therapy (RMAT) Designation Granted to AB-205 (Universal E-CEL Cell Therapy) to…
Posted: November 13, 2020 at 4:55 am
Angiocrine Bioscience Announces FDA Regenerative Medicine Advanced Therapy (RMAT) Designation Granted to AB-205
About Regenerative Medicine Advanced Therapy (RMAT) DesignationEstablished under the 21st Century Cures Act, the RMAT designation was established to facilitate development and expedite review of cell therapies and regenerative medicines intended to treat serious or life-threatening diseases or conditions. Advantages include the benefits of the FDA's Fast Track and Breakthrough Therapy Designation programs, such as early interactions with the FDA to discuss potential surrogate or intermediate endpoints to support accelerated approval.
About HDT-AHCT High-dose therapy and autologous hematopoietic cell transplantation (HDT-AHCT) is considered a standard-of-care therapy for patients with aggressive systemic Hodgkin lymphoma (HL) and non-Hodgkin lymphoma (NHL).Although efficacious and considered a potential cure, HDT-AHCT is associated with severe regimen-related toxicities (SRRT) that increase patient morbidity and risk for mortality, especially in the aging population. Effective prevention of SRRT may lead to more patients being eligible for a potential cure through HDT and stem cell transplantation.
About SRRT Consequences of Diffuse Injury to the Organ Vascular NichesThe human body is capable of renewing, healing and restoring organs.For example, the human oral-GI tract renews its lining every 3 to 7 days. Both the organ renewal and healing processes are dependent on organ stem cell vascular niches made up of stem cells, endothelial cells (cells that line blood vessels) and supportive cells.When tissues are injured, the vascular niche endothelial cells direct the stem cells, via angiocrine factor expression, to repair and restore the damaged tissue. This restorative capacity is most active during childhood and youth but starts to diminish with increasing age.HDT provided to eradicate cancer cells also cause diffuse, collateral damage to vascular niches of multiple healthy organs. In particular, the organs with the highest cell turnover (ones with most active vascular niches) are severely affected.Specifically, the oral-GI tract, dependent on constant renewal of its mucosal lining, starts to break down upon vascular niche injury.The mucosal breakdown can cause severe nausea, vomiting and diarrhea. In addition, the bacteria in the gut may escape into the circulation, resulting in patients becoming ill with endotoxemia, bacteremia or potentially lethal sepsis.HDT-related vascular niche damage can also occur in other organs resulting in severe or life-threatening complications involving the lung, heart, kidney, or the liver.Collectively, these complications are known as severe regimen-related toxicities or SRRT.SRRT can occur as frequently as 50% in lymphoma HDT-AHCT patients, with increased rate and severity in older patients.
About AB205AB-205 is a first-in-class engineered cell therapy consisting of proprietary 'universal' E-CEL (human engineered cord endothelial) cells.The AB-205 cells are intravenously administered after the completion of HDT on the same day as when the patient's own (autologous) blood stem cells are infused. AB-205 acts promptly to repair injured vascular niches of organs damaged by HDT.By repairing the vascular niches, AB-205 restores the natural process of tissue renewal, vital for organs such as oral-GI tract and the bone marrow. Successful and prompt organ restoration can prevent or reduce SRRT, an outcome that is beneficial to quality of life and cost reductive to the healthcare system.
About CIRMThe California Institute for Regenerative Medicine (CIRM) was established in November, 2004 with the passage of Proposition 71, the California Stem Cell Research and Cures Act. The statewide ballot measure provided $3 billion in funding for California universities and research institutions.With over 300 active stem cell programs in their portfolio, CIRM is the world's largest institution dedicated to stem cell research. For more information, visit http://www.cirm.ca.gov.
About Angiocrine Bioscience Inc.Angiocrine Bioscience is a clinical-stage biotechnology company developing a new and unique approach to treating serious medical conditions associated with the loss of the natural healing and regenerative capacity of the body.Based on its novel and proprietary E-CEL platform, Angiocrine is developing multiple therapies to address unmet medical needs in hematologic, musculoskeletal, gastrointestinal, soft-tissue, and degenerative/aging-related diseases.A Phase 3 registration trial is being planned for the intravenous formulation of AB-205 for the prevention of severe complications in lymphoma patients undergoing curative HDT-AHCT.This AB-205 indication is covered by the Orphan Drug Designation recently granted by the US FDA.In addition, Angiocrine is conducting clinical trials of local AB-205 injections for the treatment of: (1) rotator cuff tear in conjunction with arthroscopic repair; and, (2) non-healing perianal fistulas in post-radiation cancer patients.
For additional information, please contact:
Angiocrine Bioscience, Inc.John R. Jaskowiak(877) 784-8496[emailprotected]
SOURCE Angiocrine Bioscience, Inc.
See original here:
Angiocrine Bioscience Announces FDA Regenerative Medicine Advanced Therapy (RMAT) Designation Granted to AB-205 (Universal E-CEL Cell Therapy) to...
Posted in Cell Medicine
Comments Off on Angiocrine Bioscience Announces FDA Regenerative Medicine Advanced Therapy (RMAT) Designation Granted to AB-205 (Universal E-CEL Cell Therapy) to…
Making cancer cells more susceptible to dying Washington University School of Medicine in St. Louis – Washington University School of Medicine in St….
Posted: November 13, 2020 at 4:55 am
Visit the News Hub
Findings could aid anti-tumor drug development
Researchers at Washington University School of Medicine in St. Louis and their colleagues at Imperial College London have identified how a key protein in cancer cells changes shape to kick-start the repair of DNA damage caused by chemotherapy or radiation. Blocking this built-in repair mechanism with a drug has the potential to make chemotherapy or radiation more effective, the researchers say. The key protein is called Mec1 in yeast and ATR in humans. The video shows two Mec1 molecules bound together (one is in color on the left; the other is grey on the right). The side in color shows how the protein moves to switch between active and inactive states.
Radiation and chemotherapy are designed to kill cancer cells. But for many patients, cancer cells can survive even after being hit with high doses of chemotherapy or radiation. To make treatment more effective, scientists are focusing on ways to tweak the inner machinery of cancer cells to make them more susceptible to dying.
A team at Washington University School of Medicine in St. Louis is making headway in such efforts. The researchers have identified how a key protein in cancer cells changes shape to kick-start the repair of DNA damage caused by chemotherapy or radiation. Blocking this built-in repair mechanism with a drug has the potential to make chemotherapy or radiation more effective, according to the scientists.
The study appears Nov. 9 in the journal Nature Structural & Molecular Biology.
Because this protein is essentially the same in lower organisms as well as people, the researchers studied the version of the protein found in yeast, called Mec1. Mec1 and its human counterpart, ATR, are activated when cells are stressed. The proteins are responsible for sensing and repairing DNA damage before cells replicate to prevent that damage from being passed down to daughter cells. In some cases, this activation is good, protecting healthy cells from DNA damage that could lead to cancer. But in other cases, such as cancer therapy, doctors would like to turn these repair mechanisms off so the cancer cells are more susceptible to death by further DNA damage. In this way, cancer cells hit with radiation and chemotherapy can be destroyed more easily.
Determining the structures of both the inactive and active forms of this protein gives new insights into how that transition takes place, not just for Mec1 and ATR but for other members of the same family of proteins, said senior author Peter M. Burgers, PhD, the M. A. Brennecke Professor of Biological Chemistry. ATR inhibition is a promising anti-cancer treatment when combined with radiation or chemotherapy. A handful of ATR kinase inhibitors exists, and one called ceralasertib is being tested in phase 2 clinical trials in the U.S. Our study provides a tool for improving current ATR kinase inhibitors or designing new ones in a laboratory. Providing high-resolution structures is a critical step in the intelligent design of selective inhibitors.
To determine these structures, the Burgers lab studied yeast with various mutations in this key protein and found one mutant that forced the protein into a permanent on position. With collaborators at Imperial College London, Luke Yates, PhD and Xiaodong Zhang, PhD, the researchers then determined the structure of the protein when constantly on, at an extremely high resolution on the scale of individual atoms.
We already knew what it looks like when its off, said first author Elias A. Tannous, PhD, a senior scientist in the Burgers lab. But there was a lot of speculation about what it looks like when its turned on. How does it change its shape? Does it break in two? Does it bind to something else? We didnt know. And it was interesting to find that it changes shape like a butterfly opening its wings.
These types of proteins control many aspects of the cell, from growth and viability to replication and response to stress, Tannous said. Its the master machinery of DNA damage response responsible for accurate DNA replication. If there is any error, it tells the cell to stop. This can be good or bad depending on the situation. In future research, we can use this knowledge of the structure to learn how to fine tune the activity of this type of protein, with the goal of using this information to design more effective cancer therapies.
This work was supported in part by the Wellcome Trust and the National Institutes of Health (NIH), grant number GM118129.
Tannous EA, Yates LA, Zhang X, Burgers PM. Mechanism of auto-inhibition and activation of Mec1-ATR checkpoint kinase. Nature Structural & Molecular Biology. Nov. 9, 2020. DOI 10.1038/s41594-020-00522-0
Washington University School of Medicines 1,500 faculty physicians also are the medical staff of Barnes-Jewish and St. Louis Childrens hospitals. The School of Medicine is a leader in medical research, teaching and patient care, ranking among the top 10 medical schools in the nation by U.S. News & World Report. Through its affiliations with Barnes-Jewish and St. Louis Childrens hospitals, the School of Medicine is linked to BJC HealthCare.
Posted in Cell Medicine
Comments Off on Making cancer cells more susceptible to dying Washington University School of Medicine in St. Louis – Washington University School of Medicine in St….
Radiation Does Not Disrupt CAR T-Cell Therapy in Patients With MM, Study Finds – AJMC.com Managed Markets Network
Posted: November 13, 2020 at 4:55 am
Patients with multiple myeloma (MM) can safely undergo radiation therapy without disrupting their plan to receive chimeric antigen receptor (CAR) T-cell therapy, according to new research. The study, presented last month at the American Society for Radiation Oncologys annual meeting, was conducted online due to the coronavirus disease 2019 pandemic.
CAR T-cell therapy has increasingly been seen as a potential breakthrough treatment for patients with MM, as well as other types of cancers. The therapy involves reprogramming patients own immune cells before infusing them back into the patients, where they find and kill cancer cells.
The new study focuses on CAR T B-cell maturation antigen (CART-BCMA) treatment, which was developed at the University of Pennsylvania.
Shwetha Manjunath, MD, the studys lead author and a resident in radiation oncology at Penns Perelman School of Medicine, said in a press release that although radiation has long been used to help alleviate bone pain associated with relapsed or refractory MM, it was not known whether the therapy could be safely and effectively used in patients who would eventually go on to undergo CAR T-cell therapy.
To study this question, Manjunath and colleagues retrospectively identified 25 patients who received CART-BCMA therapy. Those patients were categorized into one of 3 groups:
The first group consisted of 4 patients. None of these patients experienced severe adverse effects like cytokine release syndrome (CRS) or grade 3 or higher gastrointestinal, infectious, liver-related, or neurological toxicities. Those patients also had lower rates of grade 4 hematologic toxicities, the authors said.
Among the 8 patients who had previous radiation, 3 experienced grade 3 or higher CRS. The remaining 13 patients, none of whom had undergone radiation in the year prior to CAR T-cell infusion, had 5 cases of grade 3 or higher CRS.
The authors found no decrease in overall survival or progression-free survival among the patients who underwent radiation.
The most important takeaway here is that bridging radiation doesnt appear to increase the risk of CRS or neurotoxicity, said Manjunath, in a press release. These patients safely received bridge radiation without it affecting the efficacy of CAR T cells or the rates of toxicity.
In fact, Manjunath said it may well be that radiation therapy actually helps the efficacy of CAR T-cell therapy.
Our work is hypothesis generating, hinting at a potential synergism between radiation and CART-BCMA therapy, which has been reported by others in the literature, she said, adding that future prospective trials of radiation and CART-BCMA are warranted to better understand the safety profile and long-term efficacy of the combination.
Reference
Manjunath SH, Cohen AD, Arscott WT, Maity A, Plastaras JP, Paydar I. Is bridging radiation (RT) safe in B cell maturation antigen-targeting chimeric antigen receptor T cells (CART-BCMA) therapy? Int J Radiat Oncol Biol Phys. 2020;108(suppl 3):S165-S166. doi:10.1016/j.ijrobp.2020.07.934
See the original post here:
Radiation Does Not Disrupt CAR T-Cell Therapy in Patients With MM, Study Finds - AJMC.com Managed Markets Network
Posted in Cell Medicine
Comments Off on Radiation Does Not Disrupt CAR T-Cell Therapy in Patients With MM, Study Finds – AJMC.com Managed Markets Network
‘Rewiring’ metabolism in insulin-producing cells may aid Type 2 diabetes treatment – University of Wisconsin-Madison
Posted: November 13, 2020 at 4:54 am
Researchers have discovered a previously unknown way that pancreatic cells decide how much insulin to secrete. It could provide a promising new target to develop drugs for boosting insulin production in people with Type 2 diabetes.
In a pair of papers recently published in Cell Metabolism, scientists from the University of WisconsinMadison and their colleagues point to an overlooked enzyme known as pyruvate kinase as the primary way pancreatic beta cells sense sugar levels and release the appropriate amount of insulin.
From several proof-of-concept experiments in rodents and on human pancreatic cells, the team found that drugs stimulating pyruvate kinase not only increase the secretion of insulin but have other metabolically protective effects in the liver, muscle and red blood cells. The findings suggest that activating pyruvate kinase could be a new way to increase insulin secretion to counter Type 2 diabetes, but more research would be required before any new treatments were available.
Too much insulin can lower blood sugar to dangerous levels, and too little insulin can lead to diabetes, says Matthew Merrins, a professor of medicine at the UW School of Medicine and Public Health who led the work. The question were asking here is: How do nutrients like glucose and amino acids turn on beta cells in the pancreas to release just the right amount of insulin?
The work was accomplished by carefully dissecting the paradoxical timing of key biochemical events in the prevailing understanding of how pancreatic beta cells respond to nutrients in the blood. The researchers point to a new, richer model to understand how this important process is controlled that resolves these inconsistencies.
For decades, scientists believed that mitochondria, the energy generators in cells, initiated insulin secretion. It was a natural explanation, because mitochondria produce the high-energy molecule ATP, in the process depleting ATPs low-energy version, ADP. The drop in ADP stimulates calcium the ultimate trigger to release stored insulin.
But the timing didnt make sense. Mitochondria are most active after insulin secretion has already begun, not before. Plus, mitochondria would stall out before exhausting enough ADP to trigger insulin secretion.
A clue to solving these apparent paradoxes came from studies in the 1980s on heart muscle cells. At the time, scientists found that the enzyme pyruvate kinase which converts sugar into energy, independently of mitochondria could also severely deplete ADP. This process happens near ADP-sensing proteins involved in insulin release in the pancreas. Maybe, Merrins team thought, the pancreas took advantage of this proximity to fine-tune the release of insulin.
In initial experiments, the researchers supplied sugar and ADP to sections of pancreatic cells containing pyruvate kinase. The enzyme gobbled up both components, depleting ADP. Because pyruvate kinase was located near the ADP-sensing protein that triggers insulin secretion, it had a big effect.
Thats one of the important concepts in our paper: the location of metabolism is critical to its function, says Merrins.
Using mouse and human pancreatic islets, the clusters of cells that release insulin, the researchers tried stimulating pyruvate kinase activity. Drugs that activate the enzyme quadrupled the release of insulin, but only when there was enough sugar around a sign that pyruvate kinase cant be forced to release too much insulin.
Pyruvate kinase doesnt change how much fuel comes into the cell, it just changes how that fuel is used, says Merrins. Drugs that active pyruvate kinase strongly boost insulin secretion without causing too much insulin release that can lead to hypoglycemia.
In all, they discovered evidence of a more complex way in which pancreatic beta cells decide when and how much insulin to release, akin to a two-cycle engine. In the first cycle, blood sugar gets processed by pyruvate kinase, depleting ADP. Mitochondria keep the process going by feeding pyruvate kinase even more material, which causes ADP levels to crash, ultimately stimulating enough calcium entry into the cell to release insulin.
In the second cycle, mitochondria switch from feeding pyruvate kinase with material to producing the high-energy molecule ATP, which is needed to fully release insulin. Then the process resets.
In the companion study, led by Merrins colleagues at Yale University, the researchers examined how pyruvate kinase activators affected metabolism in healthy and obese rats. In a series of experiments, they found that activating pyruvate kinase increased both insulin secretion and insulin sensitivity while improving sugar metabolism in liver and red blood cells. Such treatments could be helpful for people with Type 2 diabetes, who dont produce enough insulin and have dysfunctional sugar metabolism as a result.
The therapeutic idea here is we could rewire metabolism to more efficiently trigger insulin secretion while improving the function of other organs at the same time, says Merrins.
This work was supported in part by the National Institutes of Health (grants R01DK092606, R01DK110181, K08DK080142, UL1RR-0024139, P30DK045735, K01DK101683, R01DK113103, R21AG050135, R01AG062328, F32DK116542, T32AG000213, T32DK007665) and the Health Resources and Services Administration (grant T32HP10010).
The rest is here:
'Rewiring' metabolism in insulin-producing cells may aid Type 2 diabetes treatment - University of Wisconsin-Madison
Posted in Cell Medicine
Comments Off on ‘Rewiring’ metabolism in insulin-producing cells may aid Type 2 diabetes treatment – University of Wisconsin-Madison