Page 830«..1020..829830831832..840850..»

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.

See more here:
Making cancer cells more susceptible to dying Washington University School of Medicine in St. Louis - Washington University School of Medicine in St....

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

When the Language of Cells is Interrupted – UC San Diego Health

Posted: November 13, 2020 at 4:54 am

New Cell Signaling Center supports research on the most fundamental of cellular functionand development of therapeutics that target them

As Alexandra Newton took the stage late last year to give an invited lecture at the University of Dundee in Scotland, the event host introduced her as hailing from San Diego, the other top cell signaling community in the world.

Alexandra Newton (right) and members of her lab at UC San Diego School of Medicine (pictured pre-COVID-19 pandemic.

Until that moment, Newton hadnt thought of it that way. Yet it struck her as true.

We probably dont fully appreciate our own depth of expertise because its our normal, said Newton, Distinguished Professor in the Department of Pharmacology at UC San Diego School of Medicine. So, I figured if this university has a center dedicated to cell signaling, surely UC San Diego should as well.

And so Newton, with colleague Jin Zhang, professor of pharmacology, launched the Cell Signaling Center at UC San Diego. Their goal is to unite researchers passionate about understanding cell signaling, and leveraging it to treat disease.

Newton describes cell signaling as the language cells use to communicate among themselves and with the outside world. Say, a signal knocks on a cells outer door, so to speakinsulin, for example. The signal first needs to get the message inside the cell, then trigger a change in behavior, such as taking up glucose in this example, or growing, dividing or secreting.

That cascade of events is known as cell signaling, and its a language many scientists want to speak so that they can better understand the many ways it can go awry.

If I talked super-fast or way too slowly, youd have trouble understanding me, Newton said. Similarly, a change in cell signaling can lead to miscommunication, and ultimately to disease.

Within the complex language of cells, there are a few commonly used phrasesphosphorylation is one key example. In phosphorylation, enzymes called kinases add phosphate groups to proteins and other enzymes called phosphatases remove them, changing protein conformation or activity back and forth like a switch. Whether or not a phosphate group is present in a particular place and time can drastically change cell behavior.

Cell signaling malfunctions can lead to a variety of diseases. For example, a sped-up kinase in one pathway might allow cells to divide out of control, leading to cancer. A slowdown of the same kinase may contribute to untimely cell death, as might occur in neurodegenerative diseases, such as Alzheimers.

If we know what has gone wrong, we are in a better position to fix it, Newton said.

Lets say you are repeatedly inserting a wrong word in your sentences, Newton said. We need to find the wrong word and replace it with the correct wordthats what were doing with targeted therapeutics.

Often, very small changes are sufficient to disrupt the normal balance of cell signaling. If just a few atoms in one amino acid, the building block of proteins, are off in a kinase, it could cause the enzyme to start spitting out phosphate groups 30 percent faster, changing the behavior of the cell, causing Alzheimers or some other disease, and we need therapeutic drugs to slow it down.

Graduate students in Alexandra Newtons lab hold a model of Protein Kinase C (pictured pre-COVID-19 pandemic).

There are more than 500 protein kinases operating in our cells, and many current and experimental drugs work by inhibiting these enzymes. One example is the drug imatinib (brand name Gleevec), a kinase inhibitor that has been a game-changer in treating some types of leukemia.

For decades, many researchers had also attempted to develop drugs that inhibit Protein Kinase C (PKC), Newtons favorite cell signaling molecule, as a means to treat cancer. But in 2015, Newtons team reversed this 30-year paradigm when they reported evidence that PKC actually suppresses, rather than promotes, tumors. Their work implied that anti-cancer drugs would actually need to do the oppositeboost PKC activity.

PKC enzymes are the perfect example of the need to understand the complex biochemistry of our target molecules before we start targeting them in therapies, Newton said.

UC San Diegos global reputation as a leader in cell signaling research grew out of decades of discoveries.

Susan Taylor, professor in the Department of Pharmacology and Department of Chemistry and Biochemistry, is best known for solving the first crystal structure of a protein kinase (PKA, a relative of PKC) in 1991. The structure revealed how kinases are shaped, how their various pieces come together and how they perform their enzymatic reactions.

Ill never forget sitting as a group in the San Diego Supercomputer Center one afternoon, looking at a screen together and seeing PKA in 3Dit was so amazing to realize that we were the only people in the world to see the structure, and to know it was just the beginning of a big family of enzymes, Taylor said.

PKA is still considered the poster child for kinases today.

The crystal structure of Protein Kinase A (PKA), shown here, was first solved by Susan Taylor and team in 1991. Its still considered the gold standard for kinase structures today.

Taylor originally came to UC San Diego as a postdoctoral researcher in 1971, training in the lab of biochemist Nathan Kaplan. There, she worked alongside a number of talented, eager scientists, including graduate student J. Craig Venter and fellow postdoc Jack Dixon.

Venter went on to lead the effort to sequence the human genome. He is currently CEO of the J. Craig Venter Institute.

Dixon became the first to clone, express and characterize a number of phosphatases (the enzymes that do the opposite of kinasesremove phosphate groups). He also discovered the substrate of the tumor-suppressing enzyme PTEN, which shares some structural similarities with the phosphatases he studied. Dixons work suggested cell signaling pathways that could be targeted to treat cancers lacking PTEN. He is now Distinguished Professor of Pharmacology, Cellular and Molecular Medicine, and Chemistry and Biochemistry at UC San Diego, where his team is studying a largely overlooked family of secreted kinases.

In the late 1970s, Taylor also taught Dr. Brian Druker when he was an undergraduate and then a medical student at UC San Diegobefore he went on to help develop the kinase-inhibitor drug Gleevec, the first FDA-approved therapeutic that works by specifically targeting a molecular defect unique to cancer cells, making it more effective and less damaging to healthy cells.

In addition to kinases and phosphatases, UC San Diego School of Medicine is home to many experts on G-protein coupled receptors (GPCRs), molecules that span cell membranes, where they transmit messages between cells and their environments.

Like kinases, many therapeutic drugs work by influencing GPCRs. Joan Heller Brown, Distinguished Professor of Pharmacology, for example, is known for fundamental contributions to the understanding of GPCRs and how they regulate cell growth and survival in healthy and various disease states, while Dr. Paul Insel, Distinguished Professor of Pharmacology and Medicine, is an expert in the genomics of GPCRs. Silvio Gutkind, who recently succeeded Heller Brown as chair of the Department of Pharmacology, and team are developing innovative anti-cancer prevention and treatment options that work by targeting GPCRs.

Much of todays cell signaling research relies heavily on leading-edge tools that have grown out of the work of late Nobel Laureate and UC San Diego Professor Roger Tsientools that allow researchers to eavesdrop on cellular conversations.

Tsien, recruited to UC San Diego in 1989, shared the 2008 Nobel Prize in Chemistry for the discovery and application of green fluorescent protein (GFP), which allows some jellyfish to glow. While colleagues discovered and isolated the GFP gene, Tsien found ways to tweak its makeup, making it glow more brightly and consistently. Then, he created a full color palette of fluorescent proteins that scientists could use to track the molecular goings-on within a cell.

GFP and its many colorful cousins (with names like mCherry and mOrange) quickly became indispensable tools in life sciences labs around the world. Tsien, who died in 2016, often called these fluorescent tags molecular spies because they allow researchers to listen in on cellular communications, to watch molecules interact in real-time and ask questions once thought impossible.

With support from Bristol Meyers Squibb, the new Cell Signaling Center is proud to offer an Annual Roger Tsien Cell Signaling Distinguished Lecture Series in his honor.

Today, several of Tsiens trainees, including Zhang, Nathan Shaner, associate professor of neurosciences, and others are carrying on his legacy at UC San Diego School of Medicine. Shaner, original developer of the GFP-related fruit series, continues to engineer and optimize fluorescent proteins for use with newer imaging techniques. Zhang has devised a new set of fluorescent biosensors.

Our biosensors are unique in that we put in the DNA and cells assemble them for us, Zhang said. They allow us to follow a molecule of interest in a live cell, and see what it does, how, when and where.

Ultrasensitive biosensors developed by Jin Zhang and team help scientists track kinase activity in live mice.

Hardly a week goes by in which there isnt a new cell signaling study by UC San Diego School of Medicine researchers published in top scientific journals. In just the last few months:

When thinking about the future of the cell signaling field, Taylor said shes perhaps most excited about recent leaps in imaging capabilities in the past few years, especially when combined with computational and engineering expertise.

For example, researchers were (virtually) lining up to collaborate with new assistant professor Johannes Schneberg after he presented his work in quantitative 4D imaging at a recent meeting. His technology can, for example, fly viewers through the mitochondria in a live human brain organoid grown in a lab.

People across disciplines have always worked together well here at UC San Diego, even from the very beginning, and that has been enhanced by many cross-campus joint appointmentslike both Johannes Schneberg and Roger Tsien, who held faculty appointments in both the School of Medicines Department of Pharmacology and the Department of Chemistry and Biochemistry on the main campus, Taylor said.

That collaborative spirit is the basis for the new Cell Signaling Center. According to Zhang, the center will act as a hub for researchers in the field to better connect with each other, as well as a one-stop-shop for local biopharmaceutical companies and colleagues around the world looking for the types of experts and resources that UC San Diego can provide.

Together, we are taking not a sledgehammer approach to treating disease, but a precise fine-tuning of cell signaling processes to correct their mis-regulation in disease, Zhang said. Thats the basis of precision, or personalized, pharmacology, and it starts with the kind of work were doing here at UC San Diego.

Read more from the original source:
When the Language of Cells is Interrupted - UC San Diego Health

Posted in Cell Medicine | Comments Off on When the Language of Cells is Interrupted – UC San Diego Health

Researchers discover two key events that turn normal cells into cancer – Penn Today

Posted: November 13, 2020 at 4:54 am

More than 100 different cancers can arise all over the body, but two universal metabolic pathways may tie them all together, researchers from the Perelman School of Medicine report in a new study published online in Cell Metabolism. Researchers have long believed all cancers are governed by a common set of fundamental processes. Exactly what those were, however, has remained elusive.

Having a unifying mechanism could inform new therapeutic approaches to prevent normal cells from transforming into any type of tumor, be it breast, prostate, or colon, for example.

The team discovered how the transformation from a phenotypically normal cell to a cancerous one involves the enhancement of two key elements: antioxidant defense and nucleotide synthesis. Genes associated with cancer, they found, are super charging some cells to fight off oxidative stress and synthesize nucleotides, which cells need to survive and rapidly grow, respectively.

Since the early 1980s, numerous cancer genes have been identified. However, they often affect multiple cellular processes, which makes it very hard to really summarize what exactly turns cells cancerous, says senior author Xiaolu Yang, a professor of cancer biology in the Perelman School of Medicine. We took a unique approach and looked at the cellular changes driven by a particular metabolic enzyme, which turned out to be the key here. Strikingly, we found that for a phenotypically normal cell to become a cancer cell, all it needs to do is to be equipped with the extra capacity to endure oxidative stress and produce nucleotides.

Shut down these metabolic pathways, the study suggests, and the cells dont become cancerous.

Read more at Penn Medicine News.

Go here to see the original:
Researchers discover two key events that turn normal cells into cancer - Penn Today

Posted in Cell Medicine | Comments Off on Researchers discover two key events that turn normal cells into cancer – Penn Today

Common Cold Coronaviruses Tied to Less Severe COVID-19 Cases – The Scientist

Posted: November 13, 2020 at 4:54 am

There are four common cold coronaviruses that we all catch at some stage. We generate antibodies to them, but our immune memory of them fades over time, and we get re-infected.

Their names are all too easily forgottenOC43, HKU1, 229E, and NL63but our immune systems may nevertheless remember them for a time. There have been hints that exposure to these common coronaviruses might offer some protection from COVID-19, mostly by looking at signs of immune memory in blood samples taken from before the pandemic. A study in the Journal of Clinical Investigationreports the first clinical evidence linking recent endemic coronavirus infections to less severe COVID-19 and even a reduced death rate in patients.

The COVID-19 disease is actually much less severe in those patients who had documented endemic coronavirus infections.

Manish Sagar, Boston Medical Center

The authors at Boston University School of Medicine found evidence for this by poring over the medical records of thousands of patients who had visited Boston Medical Center as inpatients or outpatients, most probably for respiratory illnesses, between 2015 and 2020. Each person had been assessed for infection using a PCR test that screens for bacteria and viruses, including the four endemic coronaviruses.

In total, 15,928 patients had at least one such PCR test. Of them, 875 tested positive for an endemic coronavirus (this group was called eCoV+), while the remaining 15,053 people never had a documented coronavirus infection (termed eCoV-).

Of the entire cohort, a total of 1,812 (11.4 percent) later returned for a SARS-CoV-2 test during the initial COVID-19 surge in Boston between March 12 and June 12.

Our study is the first to examine people with known endemic coronavirus infections, and compare them to people who, as far as we know, dont have any recent documented coronavirus infections, says Manish Sagar, the lead author of the study and a virologist at Boston Medical Center.

The infection rate for SARS-CoV-2 was no different between those who had a recently recorded endemic coronavirus infection (eCoV+) and those who did not have a positive test (eCoV-). This led the authors to conclude that a recent infection with endemic coronaviruses did not keep SARS-CoV-2 at bayboth groups were just as likely to become infected with the pandemic virus.

When the researchers peered closer at the data, they observed an important difference between the two groups. The COVID-19 disease is actually much less severe in those patients who had documented endemic coronavirus infections, says Sagar. The odds of intensive care unit (ICU) admission were significantly lower in eCoV+ than in eCoV- patients, and there was a trend towards lower odds of mechanical ventilation, the authors write in their report.

The data also show that among hospitalized patients who had previous positive test results for endemic coronavirus, 4.8 percent of them died compared with 17.7 percent among those in the group without such a test result.

Local immune memory may help explain these results. Such heterotypic immunity, says immunologist Joseph Mizgerd, director of the pulmonary center at Boston University School of Medicine, occurs when immune memory is etched into the lungs and/or nose. Its common after other types of respiratory infections and might offer protection against SARS-CoV-2 if elicited by endemic coronaviruses. Although the Boston group did not measure this type of immunity in patients, they now hypothesize that local immunity gained from endemic coronaviruses helps limit lung injury during COVID-19. We are testing that in ongoing experiments, Mizgerd says by email. He adds that such cross-reactive immunity is often mediated by memory T cells, which can localize in the lung, and he notes that lung-localized heterotypic T cells can prevent severe lung infection during pneumonias caused by other types of respiratory pathogens.

If indeed prior infection does ramp up protection against SARS-CoV-2, the study could not answer how long it takes for any such benefit to taper off. Nor did the work shed light on which of the four endemic coronaviruses in particular might be offering protection against the pandemic virus. The scientists are seeking funding to expand their research and include data from other institutions.

Mizgerd and his team did not look into which immune components may be responsible for an endemic coronavirus influencing a persons immune response to SARS-CoV-2. This is something that immunologist Dennis Burton at the Scripps Research Institute in La Jolla, California, and his colleagues have investigated.

Since the start of the pandemic, they have been interested in whether pre-existing immune responses to seasonal coronaviruses could influence antibody responses to SARS-CoV-2. In a study published in September as a preprint on bioRxiv, Burton and colleagues compared serum antibodies and antibody-producing B cells from 36 donors sampled prior to the pandemic to see whether those antibodies reacted with the spike protein from the new pandemic virus. Very few antibodies from before the pandemic reacted to SARS-CoV-2, the team found. The vast majority did not bind strongly to the new virus, although they did identify one antibody that could neutralize SARS-CoV-2.

The group also detected memory B cells in blood samples from before the pandemic that were turned on by the presence of SARS-CoV-2. This activation triggered them to make antibodies that reacted against some proteins made by SARS-CoV-2. That would suggest that there is some cross reactivity there, says Burton.

A cross-protective vaccine that protects against SARS-CoV-2 plus the endemic coronaviruses would be a really great boon.

Manish Sagar, Boston Medical Center

A recent Sciencestudy reported that 5 percent of 302 adults and 43 percent of 48 children had antibodies that reacted against certain proteins produced by SARS-CoV-2. Children are more prone to common cold coronavirus infections, perhaps explaining why they might harbor such antibodies, and why they suffer less severe COVID-19 symptoms.

We do not know yet if the presence of such antibodies modifies the risk of becoming infected or the severity of disease, senior author George Kassiotis at the Francis Crick Institute in London explains by email. There are conserved parts of the S2 peptide of the spike protein, such as the fusion peptide, in most coronaviruses that are targeted by such cross-reactive and potentially cross-protective antibodies, Kassiotis notes. This may hold promise for a universal vaccine protecting against current, as well as future CoVs, the authors write in their Sciencepaper.

Kassiotis says that concerns that antibody immunity might be short-lived have now been allayed by recent studies and adds that even if antibodies fell below detectable levels, the cells that made them will still be there and will respond faster and better to re-infection.

Antibodies and B cells are part of only one aspect of our immune memory to viruses. Multiple investigations since the beginning of the pandemic have suggested that between 20 percent and 50 percent of people who had never encountered SARS-CoV-2 had T cells that nevertheless seemed to react against peptides from this virus, as noted recently in a Sciencepaper.

In another study in Nature, researchers in Singapore identified memory T cells in patients who had recovered from SARS back in 2003. These were reactive to proteins from SARS-CoV-2, supporting the idea that T cell memory from infections with human coronaviruses may play a role in the response to an infection with the new pandemic virus.

An additional study recently published in Scienceused human blood samples from before the pandemic to locate parts of SARS-CoV-2 that stimulated existing T cells. The study found a range of memory T cells that could react to both the new virus but also to the four common cold coronaviruses, again suggesting that existing T cells against common coronaviruses could play a role in the immune response to SARS-CoV-2 in some patients.

Immunologist Stanley Perlman of the University of Iowa who was not involved in any of these studies says that everybody should have memory B cells against common cold coronaviruses. We may also have memory T cells that remember these viruses and perhaps help with fighting SAR-CoV-2. However, Perlman emphasized that the implications of this for COVID-19 is still a work in progress.

Burton says he hopes to dig into a molecular understanding of the cross-reactivity of antibodies, which might help design a vaccine against not just SARS-CoV-2, but common cold coronaviruses too. These viruses usually cause mild symptoms, but not always.

A cross-protective vaccine that protects against SARS-CoV-2 plus the endemic coronaviruses would be a really great boon, says Sagar. These coronaviruses are causes of the common cold, but they are also really important causes of pneumonia, pneumonia hospitalizations, and pneumonia deaths.

Read the rest here:
Common Cold Coronaviruses Tied to Less Severe COVID-19 Cases - The Scientist

Posted in Cell Medicine | Comments Off on Common Cold Coronaviruses Tied to Less Severe COVID-19 Cases – The Scientist

Expert poses new theory on what really killed woman in Harper Woods jail – Detroit Free Press

Posted: November 13, 2020 at 4:54 am

Autoplay

Show Thumbnails

Show Captions

The Wayne County Prosecutor's Office will ask an outsidemedical examiner to review the autopsy report of a woman who died earlier this year in the Harper Woods jail.

Priscilla Slater, 38, of Detroit, died inthe Harper Woods jail on June 10. Her deathprompted protests, the firing of two police officers for altering the police report detailing the discovery of her body, and the resignation of the city's longtimemayor after he told a meeting that he understood how someone could become a white supremacist.

Priscilla Slater, 38, of Detroit, died in the Harper Woods jail on June 10.(Photo: LaTasha Slater)

Slater was arrested about 1:30 a.m. on June 9 at the Parkcrest Inn in Harper Woods, after her boyfriend, Lewis Nichols, 27, also of Detroit, allegedly fired 18 shots into the parking lot.She died the next day.

The Michigan State Police investigated Slater's death. Their findings have been turned over to Wayne County Prosecutor Kym Worthy's office, which is conducting its own investigation.

A Wayne County Medical Examiner's report reached no certain conclusion about the cause of Slater's death. In the absence of pathological findings, the report speculated that an irregular heartbeat caused by abnormally angled coronary arteriescould have resulted in Slater's sudden death.

Protesters gather outside of Harper Wood Police Department while marching through Harper Woods for Priscilla Slater on July 24, 2020. Slater was found unresponsive in a Harper Woods Police Department holding cell in June 2020.(Photo: Ryan Garza, Detroit Free Press)

But Oakland County ChiefMedical Examiner Ljubisa Dragovic, M.D., who reviewed the autopsy report at the Free Press' request, calledthat theory "nonsense."

Because of the condition of Slater's liver, Dragovic said in an interview this week, "the most likely diagnosis that one can propose, without being able to see the details ofthe microscopic slides, is that she died from alcoholwithdrawal.If she was not monitored in the cell, thatis a big problem."

The autopsy report notes that video of the jail cell appears to show Slater having a seizure about 5:10 a.m. Her body was discovered in the cellmore than seven hourslater,at12:30 p.m.,

Oakland County (Mich.) Medical Examiner Ljubisa Dragovic.(Photo: ASSOCIATED PRESS)

The report does not state a time of death,but says the seizure may have occurred as Slater died.

Dragovic saidSlater's apparent seizure was not consistentwith the heart condition the Wayne County report says killed her.

"The presenting terminal events are not in concordance with a sudden onset of arrhythmia," he said.

Worthy said Wednesday that her office had not previouslybeen aware of the possibility that Slater died of alcohol withdrawal. Alcohol withdrawal is not mentioned in the Wayne County Medical Examiner's report.

"This information was not part of any investigation," she said. "Because of this new information, we feel we need to get an independent medical examinerto review the report."

Worthy added that the need to enlist an additional expert would delay the completion of her own office's report.

Edward Jouney, an addiction specialist and assistant clinical professor in the psychiatry department at Michigan Medicine, said jails should screen people who are in custody at intake to identify substance abuse and the risk of withdrawal. He noted he was speaking generally ofjail practices.

Demonstrators march down the streets of Harper Woods on July 15, 2020, demanding to know what happened to Priscilla Slater while she was in police custody. Slater was arrested by Harper Woods police and taken into custody on June 9, 2020.(Photo: Rodney Coleman-Robinson, Detroit Free Press)

"The first thing that should be done is to take a history to see if the individual has been using any alcoholor drugs, then quantify to see how much they're using," he said. "The body acclimates to drugs or alcohol, and experiences a change in physiology. When you go into jail, you're obviously not provided with alcohol. Thebody is going to react, and go into whats called alcohol withdrawal syndrome."

Most people, he said, will survive alcohol withdrawal with a minimum of care. But some will require medical treatment, and the condition can be life-threatening.

"Some people may necessitate admission to an ICU, and can experience severe problems. If they have a history of regular alcohol use, they (jailofficials)need to regularly assess them for symptoms of alcohol withdrawal."

It's unclear whether Slater was screened for drug or alcohol use, or whether she was offered any treatment while in custody. The Wayne County Prosecutor's Office denied a Freedom of Information Act request for police reports related to Slater's arrest and time in custody, citing the ongoing investigation.

LaTasha Slater, Priscilla Slater's sister, said Wednesday that Dragovic'sexplanation makes sense. Priscilla Slater worked hard, but she played hard, LaTasha said and that often meant drinking hard.

LaTasha had rejected the idea that her sister died of a previously undetected heart abnormality. But she said Dragovic's theory provides amore plausible explanation for her sister's death.

"That makes more sense to me thananything Ive heard about the whole situation," LaTasha said.

Nancy Kaffer is a columnist and member of the Free Press editorial board. She has covered local, state and national politics for two decades. Contact: nkaffer@freepress.com. Become a subscriber.

Read or Share this story: https://www.freep.com/story/news/local/michigan/wayne/2020/11/12/priscilla-slater-autopsy-jail-harper-woods/6251487002/

Read more:
Expert poses new theory on what really killed woman in Harper Woods jail - Detroit Free Press

Posted in Cell Medicine | Comments Off on Expert poses new theory on what really killed woman in Harper Woods jail – Detroit Free Press

Predicting the risk of pT3a stage in cT1 clear cell renal cell carcinoma. – UroToday

Posted: November 13, 2020 at 4:54 am

We hypothesized that pT3a stage at nephrectomy can be accurately predicted in cT1N0M0 clear cell-renal cell carcinoma (cc-RCC) patients. Of 236 patients, treated with either partial or radical nephrectomy (2005-2019), 25 (10. 6%) harbored pT3a stage. Multivariable logistic regression models predicting pT3a were fitted using age, tumor size, tumor location and exophytic rate. The new model was 81% accurate. In calibration plots, minimal departures from ideal prediction were recorded. In decision curve analyses, a net-benefit throughout all threshold probabilities was recorded relative to the treat-all or treat-none strategies. Using a probability cut-off of 21% for presence of pT3a stage, 38 patients (16.1%) were identified, in whom pT3a rate was 36.8%. Conversely, in 198 patients (83.9%) below that cut-off, the rate of pT3a was 5.6%. Alternative user-defined cut-offs may be selected. The new model more accurately identifies a subgroup of cT1N0M0 cc-RCC patients with substantially higher risk of pT3a stage than average.

European journal of surgical oncology : the journal of the European Society of Surgical Oncology and the British Association of Surgical Oncology. 2020 Nov 05 [Epub ahead of print]

Luigi Nocera, Lara F Stolzenbach, Claudia Coll Ruvolo, Mike Wenzel, Zhe Tian, Giuseppe Rosiello, Carlo A Bravi, Luigi Candela, Giuseppe Basile, Alessandro Larcher, Shahrokh F Shariat, Roberto Bertini, Umberto Capitanio, Andrea Salonia, Francesco Montorsi, Alberto Briganti, Pierre I Karakiewicz

Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montreal Health Center, Montreal, Quebec, Canada; Division of Experimental Oncology/Unit of Urology, URI, Urological Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy. Electronic address: ., Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montreal Health Center, Montreal, Quebec, Canada; Martini-Klinik Prostate Cancer Center, University Hospital Hamburg-Eppendorf, Hamburg, Germany., Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montreal Health Center, Montreal, Quebec, Canada; Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples Federico II, Italy., Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montreal Health Center, Montreal, Quebec, Canada; Department of Urology, University Hospital Frankfurt, Frankfurt am Main, Germany., Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montreal Health Center, Montreal, Quebec, Canada., Division of Experimental Oncology/Unit of Urology, URI, Urological Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy., Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Departments of Urology, Weill Cornell Medical College, New York, NY, USA; Department of Urology, University of Texas Southwestern, Dallas, TX, USA; Department of Urology, Second Faculty of Medicine, Charles University, Prag, Czech Republic; Institute for Urology and Reproductive Health, I.M. Sechenov First Moscow State Medical University, Moscow, Russia; Division of Urology, Department of Special Surgery, Jordan University Hospital, The University of Jordan, Amman, Jordan.

PubMed http://www.ncbi.nlm.nih.gov/pubmed/33168336

More:
Predicting the risk of pT3a stage in cT1 clear cell renal cell carcinoma. - UroToday

Posted in Cell Medicine | Comments Off on Predicting the risk of pT3a stage in cT1 clear cell renal cell carcinoma. – UroToday

The story of mRNA: From a loose idea to a tool that may help curb Covid – STAT

Posted: November 11, 2020 at 1:57 pm

ANDOVER, Mass. The liquid that many hope could help end the Covid-19 pandemic is stored in a nondescript metal tank in a manufacturing complex owned by Pfizer, one of the worlds biggest drug companies. There is nothing remarkable about the container, which could fit in a walk-in closet, except that its contents could end up in the worlds first authorized Covid-19 vaccine.

Pfizer, a 171-year-old Fortune 500 powerhouse, has made a billion-dollar bet on that dream. So has a brash, young rival just 23 miles away in Cambridge, Mass. Moderna, a 10-year-old biotech company with billions in market valuation but no approved products, is racing forward with a vaccine of its own. Its new sprawling drug-making facility nearby is hiring workers at a fast clip in the hopes of making history and a lot of money.

In many ways, the companies and their leaders couldnt be more different. Pfizer, working with a little-known German biotech called BioNTech, has taken pains for much of the year to manage expectations. Moderna has made nearly as much news for its stream of upbeat press releases, executives stock sales, and spectacular rounds of funding as for its science.

advertisement

Each is well-aware of the other in the race to be first.

But what the companies share may be bigger than their differences: Both are banking on a genetic technology that has long held huge promise but has so far run into biological roadblocks. It is called synthetic messenger RNA, an ingenious variation on the natural substance that directs protein production in cells throughout the body. Its prospects have swung billions of dollars on the stock market, made and imperiled scientific careers, and fueled hopes that it could be a breakthrough that allows society to return to normalcy after months living in fear.

advertisement

Both companies have been frequently name-checked by President Trump. Pfizer reported strong, but preliminary, data on Monday, and Moderna is expected to follow suit soon with a glimpse of its data. Both firms hope these preliminary results will allow an emergency deployment of their vaccines millions of doses likely targeted to frontline medical workers and others most at risk of Covid-19.

There are about a dozen experimental vaccines in late-stage clinical trials globally, but the ones being tested by Pfizer and Moderna are the only two that rely on messenger RNA.

For decades, scientists have dreamed about the seemingly endless possibilities of custom-made messenger RNA, or mRNA.

Researchers understood its role as a recipe book for the bodys trillions of cells, but their efforts to expand the menu have come in fits and starts. The concept: By making precise tweaks to synthetic mRNA and injecting people with it, any cell in the body could be transformed into an on-demand drug factory.

But turning scientific promise into medical reality has been more difficult than many assumed. Although relatively easy and quick to produce compared to traditional vaccine-making, no mRNA vaccine or drug has ever won approval.

Even now, as Moderna and Pfizer test their vaccines on roughly 74,000 volunteers in pivotal vaccine studies, many experts question whether the technology is ready for prime time.

I worry about innovation at the expense of practicality, Peter Hotez, dean of the National School of Tropical Medicine at Baylor College of Medicine and an authority on vaccines, said recently. The U.S. governments Operation Warp Speed program, which has underwritten the development of Modernas vaccine and pledged to buy Pfizers vaccine if it works, is weighted toward technology platforms that have never made it to licensure before.

Whether mRNA vaccines succeed or not, their path from a gleam in a scientists eye to the brink of government approval has been a tale of personal perseverance, eureka moments in the lab, soaring expectations and an unprecedented flow of cash into the biotech industry.

It is a story that began three decades ago, with a little-known scientist who refused to quit.

Before messenger RNA was a multibillion-dollar idea, it was a scientific backwater. And for the Hungarian-born scientist behind a key mRNA discovery, it was a career dead-end.

Katalin Karik spent the 1990s collecting rejections. Her work, attempting to harness the power of mRNA to fight disease, was too far-fetched for government grants, corporate funding, and even support from her own colleagues.

It all made sense on paper. In the natural world, the body relies on millions of tiny proteins to keep itself alive and healthy, and it uses mRNA to tell cells which proteins to make. If you could design your own mRNA, you could, in theory, hijack that process and create any protein you might desire antibodies to vaccinate against infection, enzymes to reverse a rare disease, or growth agents to mend damaged heart tissue.

In 1990, researchers at the University of Wisconsin managed to make it work in mice. Karik wanted to go further.

The problem, she knew, was that synthetic RNA was notoriously vulnerable to the bodys natural defenses, meaning it would likely be destroyed before reaching its target cells. And, worse, the resulting biological havoc might stir up an immune response that could make the therapy a health risk for some patients.

It was a real obstacle, and still may be, but Karik was convinced it was one she could work around. Few shared her confidence.

Every night I was working: grant, grant, grant, Karik remembered, referring to her efforts to obtain funding. And it came back always no, no, no.

By 1995, after six years on the faculty at the University of Pennsylvania, Karik got demoted. She had been on the path to full professorship, but with no money coming in to support her work on mRNA, her bosses saw no point in pressing on.

She was back to the lower rungs of the scientific academy.

Usually, at that point, people just say goodbye and leave because its so horrible, Karik said.

Theres no opportune time for demotion, but 1995 had already been uncommonly difficult. Karik had recently endured a cancer scare, and her husband was stuck in Hungary sorting out a visa issue. Now the work to which shed devoted countless hours was slipping through her fingers.

I thought of going somewhere else, or doing something else, Karik said. I also thought maybe Im not good enough, not smart enough. I tried to imagine: Everything is here, and I just have to do better experiments.

In time, those better experiments came together. After a decade of trial and error, Karik and her longtime collaborator at Penn Drew Weissman, an immunologist with a medical degree and Ph.D. from Boston University discovered a remedy for mRNAs Achilles heel.

The stumbling block, as Kariks many grant rejections pointed out, was that injecting synthetic mRNA typically led to that vexing immune response; the body sensed a chemical intruder, and went to war. The solution, Karik and Weissman discovered, was the biological equivalent of swapping out a tire.

Every strand of mRNA is made up of four molecular building blocks called nucleosides. But in its altered, synthetic form, one of those building blocks, like a misaligned wheel on a car, was throwing everything off by signaling the immune system. So Karik and Weissman simply subbed it out for a slightly tweaked version, creating a hybrid mRNA that could sneak its way into cells without alerting the bodys defenses.

That was a key discovery, said Norbert Pardi, an assistant professor of medicine at Penn and frequent collaborator. Karik and Weissman figured out that if you incorporate modified nucleosides into mRNA, you can kill two birds with one stone.

That discovery, described in a series of scientific papers starting in 2005, largely flew under the radar at first, said Weissman, but it offered absolution to the mRNA researchers who had kept the faith during the technologys lean years. And it was the starter pistol for the vaccine sprint to come.

And even though the studies by Karik and Weissman went unnoticed by some, they caught the attention of two key scientists one in the United States, another abroad who would later help found Moderna and Pfizers future partner, BioNTech.

Derrick Rossi, a native of Toronto who rooted for the Maple Leafs and sported a soul patch, was a 39-year-old postdoctoral fellow in stem cell biology at Stanford University in 2005 when he read the first paper. Not only did he recognize it as groundbreaking, he now says Karik and Weissman deserve the Nobel Prize in chemistry.

If anyone asks me whom to vote for some day down the line, I would put them front and center, he said. That fundamental discovery is going to go into medicines that help the world.

But Rossi didnt have vaccines on his mind when he set out to build on their findings in 2007 as a new assistant professor at Harvard Medical School running his own lab.

He wondered whether modified messenger RNA might hold the key to obtaining something else researchers desperately wanted: a new source of embryonic stem cells.

In a feat of biological alchemy, embryonic stem cells can turn into any type of cell in the body. That gives them the potential to treat a dizzying array of conditions, from Parkinsons disease to spinal cord injuries.

But using those cells for research had created an ethical firestorm because they are harvested from discarded embryos.

Rossi thought he might be able to sidestep the controversy. He would use modified messenger molecules to reprogram adult cells so that they acted like embryonic stem cells.

He asked a postdoctoral fellow in his lab to explore the idea. In 2009, after more than a year of work, the postdoc waved Rossi over to a microscope. Rossi peered through the lens and saw something extraordinary: a plate full of the very cells he had hoped to create.

Rossi excitedly informed his colleague Timothy Springer, another professor at Harvard Medical School and a biotech entrepreneur. Recognizing the commercial potential, Springer contacted Robert Langer, the prolific inventor and biomedical engineering professor at the Massachusetts Institute of Technology.

On a May afternoon in 2010, Rossi and Springer visited Langer at his laboratory in Cambridge. What happened at the two-hour meeting and in the days that followed has become the stuff of legend and an ego-bruising squabble.

Langer is a towering figure in biotechnology and an expert on drug-delivery technology. At least 400 drug and medical device companies have licensed his patents. His office walls display many of his 250 major awards, including the Charles Stark Draper Prize, considered the equivalent of the Nobel Prize for engineers.

As he listened to Rossi describe his use of modified mRNA, Langer recalled, he realized the young professor had discovered something far bigger than a novel way to create stem cells. Cloaking mRNA so it could slip into cells to produce proteins had a staggering number of applications, Langer thought, and might even save millions of lives.

I think you can do a lot better than that, Langer recalled telling Rossi, referring to stem cells. I think you could make new drugs, new vaccines everything.

Langer could barely contain his excitement when he got home to his wife.

This could be the most successful company in history, he remembered telling her, even though no company existed yet.

Three days later Rossi made another presentation, to the leaders of Flagship Ventures. Founded and run by Noubar Afeyan, a swaggering entrepreneur, the Cambridge venture capital firm has created dozens of biotech startups. Afeyan had the same enthusiastic reaction as Langer, saying in a 2015 article in Nature that Rossis innovation was intriguing instantaneously.

Within several months, Rossi, Langer, Afeyan, and another physician-researcher at Harvard formed the firm Moderna a new word combining modified and RNA.

Springer was the first investor to pledge money, Rossi said. In a 2012 Moderna news release, Afeyan said the firms promise rivals that of the earliest biotechnology companies over 30 years ago adding an entirely new drug category to the pharmaceutical arsenal.

But although Moderna has made each of the founders hundreds of millions of dollars even before the company had produced a single product Rossis account is marked by bitterness. In interviews with the Globe in October, he accused Langer and Afeyan of propagating a condescending myth that he didnt understand his discoverys full potential until they pointed it out to him.

Its total malarkey, said Rossi, who ended his affiliation with Moderna in 2014. Im embarrassed for them. Everybody in the know actually just shakes their heads.

Rossi said that the slide decks he used in his presentation to Flagship noted that his discovery could lead to new medicines. Thats the thing Noubar has used to turn Flagship into a big company, and he says it was totally his idea, Rossi said.

Afeyan, the chair of Moderna, recently credited Rossi with advancing the work of the Penn scientists. But, he said, that only spurred Afeyan and Langer to ask the question, Could you think of a code molecule that helps you make anything you want within the body?

Langer, for his part, told STAT and the Globe that Rossi made an important finding but had focused almost entirely on the stem cell thing.

Despite the squabbling that followed the birth of Moderna, other scientists also saw messenger RNA as potentially revolutionary.

In Mainz, Germany, situated on the left bank of the Rhine, another new company was being formed by a married team of researchers who would also see the vast potential for the technology, though vaccines for infectious diseases werent on top of their list then.

A native of Turkey, Ugur Sahin moved to Germany after his father got a job at a Ford factory in Cologne. His wife, zlem Treci had, as a child, followed her father, a surgeon, on his rounds at a Catholic hospital. She and Sahin are physicians who met in 1990 working at a hospital in Saarland.

The couple have long been interested in immunotherapy, which harnesses the immune system to fight cancer and has become one of the most exciting innovations in medicine in recent decades. In particular, they were tantalized by the possibility of creating personalized vaccines that teach the immune system to eliminate cancer cells.

Both see themselves as scientists first and foremost. But they are also formidable entrepreneurs. After they co-founded another biotech, the couple persuaded twin brothers who had invested in that firm, Thomas and Andreas Strungmann, to spin out a new company that would develop cancer vaccines that relied on mRNA.

That became BioNTech, another blended name, derived from Biopharmaceutical New Technologies. Its U.S. headquarters is in Cambridge. Sahin is the CEO, Treci the chief medical officer.

We are one of the leaders in messenger RNA, but we dont consider ourselves a messenger RNA company, said Sahin, also a professor at the Mainz University Medical Center. We consider ourselves an immunotherapy company.

Like Moderna, BioNTech licensed technology developed by the Pennsylvania scientist whose work was long ignored, Karik, and her collaborator, Weissman. In fact, in 2013, the company hired Karik as senior vice president to help oversee its mRNA work.

But in their early years, the two biotechs operated in very different ways.

In 2011, Moderna hired the CEO who would personify its brash approach to the business of biotech.

Stphane Bancel was a rising star in the life sciences, a chemical engineer with a Harvard MBA who was known as a businessman, not a scientist. At just 34, he became CEO of the French diagnostics firm BioMrieux in 2007 but was wooed away to Moderna four years later by Afeyan.

Moderna made a splash in 2012 with the announcement that it had raised $40 million from venture capitalists despite being years away from testing its science in humans. Four months later, the British pharmaceutical giant AstraZeneca agreed to pay Moderna a staggering $240 million for the rights to dozens of mRNA drugs that did not yet exist.

The biotech had no scientific publications to its name and hadnt shared a shred of data publicly. Yet it somehow convinced investors and multinational drug makers that its scientific findings and expertise were destined to change the world. Under Bancels leadership, Moderna would raise more than $1 billion in investments and partnership funds over the next five years.

Modernas promise and the more than $2 billion it raised before going public in 2018 hinged on creating a fleet of mRNA medicines that could be safely dosed over and over. But behind the scenes the companys scientists were running into a familiar problem. In animal studies, the ideal dose of their leading mRNA therapy was triggering dangerous immune reactions the kind for which Karik had improvised a major workaround under some conditions but a lower dose had proved too weak to show any benefits.

Moderna had to pivot. If repeated doses of mRNA were too toxic to test in human beings, the company would have to rely on something that takes only one or two injections to show an effect. Gradually, biotechs self-proclaimed disruptor became a vaccines company, putting its experimental drugs on the back burner and talking up the potential of a field long considered a loss-leader by the drug industry.

Meanwhile BioNTech has often acted like the anti-Moderna, garnering far less attention.

In part, that was by design, said Sahin. For the first five years, the firm operated in what Sahin called submarine mode, issuing no news releases, and focusing on scientific research, much of it originating in his university lab. Unlike Moderna, the firm has published its research from the start, including about 150 scientific papers in just the past eight years.

In 2013, the firm began disclosing its ambitions to transform the treatment of cancer and soon announced a series of eight partnerships with major drug makers. BioNTech has 13 compounds in clinical trials for a variety of illnesses but, like Moderna, has yet to get a product approved.

When BioNTech went public last October, it raised $150 million, and closed with a market value of $3.4 billion less than half of Modernas when it went public in 2018.

Despite his role as CEO, Sahin has largely maintained the air of an academic. He still uses his university email address and rides a 20-year-old mountain bicycle from his home to the office because he doesnt have a drivers license.

Then, late last year, the world changed.

Shortly before midnight, on Dec. 30, the International Society for Infectious Diseases, a Massachusetts-based nonprofit, posted an alarming report online. A number of people in Wuhan, a city of more than 11 million people in central China, had been diagnosed with unexplained pneumonia.

Chinese researchers soon identified 41 hospitalized patients with the disease. Most had visited the Wuhan South China Seafood Market. Vendors sold live wild animals, from bamboo rats to ostriches, in crowded stalls. That raised concerns that the virus might have leaped from an animal, possibly a bat, to humans.

After isolating the virus from patients, Chinese scientists on Jan. 10 posted online its genetic sequence. Because companies that work with messenger RNA dont need the virus itself to create a vaccine, just a computer that tells scientists what chemicals to put together and in what order, researchers at Moderna, BioNTech, and other companies got to work.

A pandemic loomed. The companies focus on vaccines could not have been more fortuitous.

Moderna and BioNTech each designed a tiny snip of genetic code that could be deployed into cells to stimulate a coronavirus immune response. The two vaccines differ in their chemical structures, how the substances are made, and how they deliver mRNA into cells. Both vaccines require two shots a few weeks apart.

The biotechs were competing against dozens of other groups that employed varying vaccine-making approaches, including the traditional, more time-consuming method of using an inactivated virus to produce an immune response.

Moderna was especially well-positioned for this moment.

Forty-two days after the genetic code was released, Modernas CEO Bancel opened an email on Feb. 24 on his cellphone and smiled, as he recalled to the Globe. Up popped a photograph of a box placed inside a refrigerated truck at the Norwood plant and bound for the National Institute of Allergy and Infectious Diseases in Bethesda, Md. The package held a few hundred vials, each containing the experimental vaccine.

Moderna was the first drug maker to deliver a potential vaccine for clinical trials. Soon, its vaccine became the first to undergo testing on humans, in a small early-stage trial. And on July 28, it became the first to start getting tested in a late-stage trial in a scene that reflected the firms receptiveness to press coverage.

The first volunteer to get a shot in Modernas late-stage trial was a television anchor at the CNN affiliate in Savannah, Ga., a move that raised eyebrows at rival vaccine makers.

Along with those achievements, Moderna has repeatedly stirred controversy.

On May 18, Moderna issued a press release trumpeting positive interim clinical data. The firm said its vaccine had generated neutralizing antibodies in the first eight volunteers in the early-phase study, a tiny sample.

But Moderna didnt provide any backup data, making it hard to assess how encouraging the results were. Nonetheless, Modernas share price rose 20% that day.

Some top Moderna executives also drew criticism for selling shares worth millions, including Bancel and the firms chief medical officer, Tal Zaks.

In addition, some critics have said the government has given Moderna a sweetheart deal by bankrolling the costs for developing the vaccine and pledging to buy at least 100 million doses, all for $2.48 billion.

That works out to roughly $25 a dose, which Moderna acknowledges includes a profit.

In contrast, the government has pledged more than $1 billion to Johnson & Johnson to manufacture and provide at least 100 million doses of its vaccine, which uses different technology than mRNA. But J&J, which collaborated with Beth Israel Deaconess Medical Centers Center for Virology and Vaccine Research and is also in a late-stage trial, has promised not to profit off sales of the vaccine during the pandemic.

Over in Germany, Sahin, the head of BioNTech, said a Lancet article in January about the outbreak in Wuhan, an international hub, galvanized him.

We understood that this would become a pandemic, he said.

The next day, he met with his leadership team.

I told them that we have to deal with a pandemic which is coming to Germany, Sahin recalled.

He also realized he needed a strong partner to manufacture the vaccine and thought of Pfizer. The two companies had worked together before to try to develop mRNA influenza vaccines. In March, he called Pfizers top vaccine expert, Kathrin Jansen.

Go here to read the rest:
The story of mRNA: From a loose idea to a tool that may help curb Covid - STAT

Posted in Pennsylvania Stem Cells | Comments Off on The story of mRNA: From a loose idea to a tool that may help curb Covid – STAT

It’s time for Kentucky to talk to expectant parents about benefits of cord blood banking – Courier Journal

Posted: November 11, 2020 at 1:53 pm

Paula Grisanti, Opinion contributor Published 6:20 a.m. ET Nov. 9, 2020

This year, Nov.17 has been designated World Cord Blood Day, an annual event to raise awareness for the life-saving benefits of cord blood transplants while educating parents, health professionals and the general public about the need to preserve these precious cells.

Cord blood transplants are being used to treat more than 80 different diseases and conditions including blood cancers like leukemia and lymphoma, neuroblastoma (the most common cancer in infants), bone-marrow failure disorders, inherited blood disorders and rare immune system disorders. They are also showing new promise for conditions that have never had treatment options, like autism and brain injury.

The first cord blood stem cell transplant, an international effort between physicians in the U.S. and Europe, was performed in France in 1988. Stem cells collected from a newborns umbilical cord blood were used to save the life of her brother, a 5-year-old with Fanconi Anemia. Since then, there have been more than 40,000 cord blood transplants performed worldwide.

Now standard of care for cancers of the blood and a host of other life-threatening diseases, blood forming stem cells for transplantation can be collected from bone marrow, circulating bloodor a newborn babys umbilical cord blood. Some experts believe cord blood contains nearly 10 times the number of stem cells found in bone marrow.

Because umbilical cord stem cells are less mature than adult bone marrow stem cells, they are also less likely to be rejected and can be used when there isnt a perfect match.

Between these threeoptions, the easiest collection by far is from umbilical cord and placental tissue after a baby is born and the umbilical cord has been cut, at no risk to mother or child, in a process that typically takes 5 to 10 minutes. The cells are then frozen in liquid nitrogen and can be stored indefinitely in private or public cord blood banks.

To store your babys cord blood for use by your child and your family only, you make arrangements with a private cord blood bank ahead of delivery to collect and store the cells; the cost to you includes a collection fee of $1,500 to $2,000 and an annual storage fee of $100 to $125.

If you cant afford or dont wish to save your babys cord blood stem cells, you can donate them to a public cord blood bank at no cost to you or your family.

Its the equivalent of registering these potentially life-saving cells with the national bone marrow registry; they will be available to the families of other children who need to find a bone marrow match after a devastating diagnosis. Without information and education, however, 95% of all cord blood is discarded as medical waste.

Right now, there is no public cord banking option in Kentucky, although public cord blood banking is highly recommended by both the American Academy of Pediatrics (AAP) and the American Medical Association (AMA). There are fewer than 25 public or hybrid cord blood banks in the U.S., many limited to a specific geographic area. None of them include Kentucky.

The chances of finding a bone marrow match in your family are only about 25%, making the bone marrow and umbilical cord blood registries a lifeline in desperate situations. Odds are worse for African Americans and other ethnic minorities who are underrepresented on the registry and ethnicity matters in a bone marrow transplant.

Donating cord blood cells to a public bank adds to the library of cells that may save someones life and increases the chance of a match for all of us. Who benefits most? Children, patients with rare human leukocyte antigen (HLA) types and ethnic minorities.

We need to do two things: Make public cord blood banking an option in the commonwealth of Kentucky, and then encourage conversations between health care providers and expectant parents about preserving these life-saving cells.

There are 28 states with legislation that ask or mandates physicians to talk to expectant parents about cord blood banking. Kentucky is not one of them, but most of our surrounding states have such legislation in place.

Through a long-standing relationship between the National Stem Cell Foundationand world-renowned cord blood expert Dr. Joanne Kurtzberg, we have a path forward for training hospitals and collecting cells for storage at the Carolinas Cord Blood Bank (CCBB), one of the largest public cord blood banks in the world. Dr. Kurtzberg directs both the Pediatric Blood and Marrow Transplant (PBMT) program at Duke University and the CCBB.

She performed the worlds first unrelated cord blood transplant in 1993, paving the way for this now routine source of donor cells for children who need a bone marrow transplant and dont have a matched donor. She established the CCBB in 1998.

Paula Grisanti is CEO of the National Stem Cell Foundation.(Photo: provided)

While weve initiated discussions between Louisville hospital systems and the CCBB, we need to begin the process of education for parents, nursing and medical school students, residents, midwives, practicing OB-GYNs and the general public.

What a waste to discard these life-saving cells the future of current and developing therapies for disabling and life-threatening diseases depends on our ability to make sure that doesnt happen.

Dr. Paula Grisanti is CEO and a founding member of the National Stem Cell Foundation, headquartered in Louisville, Kentucky. She holds a D.M.D. and MBA from the University of Louisville and has been actively involved in new venture start-ups for most of her career.

Read or Share this story: https://www.courier-journal.com/story/opinion/2020/11/09/expectant-parents-need-to-know-benefits-of-cord-blood-banking/6064540002/

See the original post:
It's time for Kentucky to talk to expectant parents about benefits of cord blood banking - Courier Journal

Posted in Kentucky Stem Cells | Comments Off on It’s time for Kentucky to talk to expectant parents about benefits of cord blood banking – Courier Journal

Page 830«..1020..829830831832..840850..»