Tag Archives: Clinical

Scientists tie third clinical trial death to experimental Alzheimer’s drug | Science

As enthusiasm mounts for a new experimental antibody that appears to slow cognitive decline in some Alzheimer’s patients, a third death linked to the drug during its clinical testing may amplify concerns about its safety. Science has obtained medical records showing a 79-year-old Florida woman participating in an ongoing trial of the antibody died in mid-September after experiencing extensive brain swelling and bleeding, as well as seizures. Multiple neuroscientists who reviewed the records at Science’s request believe her death was likely caused by the antibody, lecanemab.

“The brain swelling and the microhemorrhages … could be a serious side effect of the study medication,” and should be evaluated by trial investigators, says Ellis van Etten, a neuroscientist and neurologist at Leiden University.

The clinical trial’s sponsor, Japanese biotech Eisai Co., did not divulge the fatality at a major Alzheimer’s meeting last month where it detailed data from lecanemab’s phase 3 trial. The death came in an extension of that trial, but some scientists say it should have still been noted at the conference. “The failure of Eisai and [lecanemab codeveloper] Biogen to disclose this case … is concerning and undermines my confidence that the reported safety data is complete,” says Vanderbilt University neurologist and neuroscientist Matthew Schrag, who also reviewed the woman’s records.

The newly revealed death comes on top on other reports of serious brain bleeding and swelling in the core clinical trial and two other deaths in the extension phase—the first reported by STAT and the second by Science—that some scientists have linked to lecanemab.

Eisai, which attributed the prior fatalities and brain injuries to factors unrelated to lecanemab, declined to comment on the Florida woman’s death, citing patient privacy concerns. “All serious events, including fatalities, are reported to Eisai and considered in our evaluation of the study,” a company spokesperson said in a written statement to Science. “This information is provided to the FDA [Food and Drug Administration] and other regulatory authorities,” as well as independent review boards for the study. 

The spokesperson added that the age and medical condition of any trial participants should be considered when evaluating a death. The Florida woman, however, had no obvious health problems, other than her signs of early Alzheimer’s disease, according to her medical records.

Eisai has reported 13 deaths in the core clinical trial, which involved about 1800 people. Deaths are expected given the study population’s age and health, and the company says the numbers were similar in the groups receiving lecanemab and the placebo. But it has not made public the details of each death, so in most cases scientists have been unable to independently assess whether lecanemab contributed to the fatalities.

Lecanemab is one of several experimental Alzheimer’s drugs that target beta amyloid, the protein that builds up in the brains of people with the disease. Many in the field believe it is responsible for the brain cell death that robs people of memories and ultimately kills them, although deposits of the protein are also found in brains of healthy people.

Amyloid-seeking antibodies often cause brain swelling and bleeding, a condition known as amyloid-related imaging abnormalities (ARIA) because it is diagnosed through brain imaging. “We need a name change … because these are not just imaging abnormalities, as this case illustrates,” says Boston University neurologist and neuroscientist Andreas Charidimou, who examined the woman’s records for Science. “It’s a real clinical syndrome, which can be fatal.”

Although lecanemab targets a soluble version of beta amyloid, it also binds to the extracellular beta amyloid “plaques,” considered a hallmark of Alzheimer’s. About half of Alzheimer’s patients have a condition called cerebral amyloid angiopathy (CAA), in which beta amyloid plaques replace the smooth muscle of blood vessel walls. When antibodies such as lecanemab strip away those plaques, blood vessels can become inflamed and weakened, increasing a person’s susceptibility to ARIA.

In the two previous deaths tied to lecanemab, neurologists say the patients’ use of anticoagulants worsened brain swelling and bleeding. The Florida woman was given a minimal course of the anticoagulant heparin after being hospitalized, but several neurologists discounted it as a contributing factor to her sudden problems and ultimate death.

Whether the woman received infusions of the antibody or a placebo during the core 18-month trial is unclear. But she did get the drug over 6 weeks in the extension phase—in which any participant can opt for treatment. Before the extension trial started, a brain scan revealed signs of a few microhemorrhages, but they were not serious enough to rule her out of the trial.

One of the Florida woman’s daughters provided the medical documents to Science and authorized their review by others. To protect the family’s privacy, Science is withholding the names of the patient, the daughter, a friend who served as the woman’s helper during the study, and the clinical trial site where the patient received lecanemab.

A textbook case

The woman’s friend described a harrowing series of events that began with the patient’s first infusion of the antibody as part of the extension trial, in August. “She was so tired. She … didn’t get out of bed for 2 days other than to maybe eat a yogurt or go to the bathroom,” the friend says. A couple of weeks later, after the second infusion, the woman complained of severe headaches, “couldn’t complete sentences,” and increasingly felt confused about everyday matters, her friend recalls.

At a restaurant on 14 September, the woman experienced what seemed like a stroke. She was rushed to the hospital, where her friend informed doctors that the woman was taking the experimental drug. Seizures began, causing her to thrash her arms and legs, requiring restraints for her safety.

Before a Florida woman received lecanemab in an extension phase of a clinical trial, an MRI scan of her brain (left) had a few microhemorrhages—tiny bleeds (dark spots, examples marked by arrows). Afterward (right), dozens of microhemorrhages were obvious (examples marked by arrows).Provided anonymously to Science

Brain scans showed dozens of areas of bleeding and brain swelling so extensive that the characteristic folds of the cerebral cortex were “merged and squashed” in substantial parts of her brain, Charidimou says. He calls it “a textbook case of severe ARIA, both the clinical presentation and the imaging manifestations.” Given the absence of other potential causes for brain damage indicated in the medical records, he adds, lecanemab almost certainly was the culprit.

The hospital records show the clinical trial site investigator, contacted after the woman was hospitalized, suspected ARIA and urged treatment with steroids—which the physicians tried without significant benefit. She began to suffer from multiorgan failure and pneumonia, and died 5 days after being admitted.

“The patient had extensive swelling of her brain with some small areas of bleeding which caused her to have a seizure and ultimately to die,” says Schrag, who is a CAA specialist. “I am confident this was a side effect from lecanemab.”

Eric Smith, a neurologist at the University of Calgary who also reviewed the case materials, agrees the drug likely caused the death. He previously consulted for Eisai partner Biogen and was an investigator for the two companies’ other antiamyloid drug aducanumab (marketed as Aduhelm), which won FDA marketing approval last year.

The family has arranged for an autopsy, which could confirm that the woman had CAA and clarify the antibody’s role in her death, but it has not been completed, the daughter says. The Eisai spokesperson said the company “is thorough and proactive” in its efforts to obtain any safety information, including autopsy results for trial participants.

A lack of consensus

The deaths linked to the antibody cast a pall over recent trial results largely seen as hopeful. Eisai has reported that lecanemab slowed the rate of cognitive decline among early Alzheimer’s patients by an average of 27% over 18 months, a statistically significant effect. Neurologists differ on whether that benefit would be noticeable to many patients or caregivers, and some large subgroups in the trial, including women and people under age 65, did not benefit to a statistically significant threshold.

Still, the trial represented the most favorable results for any antiamyloid therapy so far and has prompted calls from some Alzheimer’s scientists and patient groups for FDA to quickly green-light the drug.

Earlier this month, a lecanemab “consensus statement”—whose initial signers have worked as consultants to Eisai or Biogen or have conducted research for the recent lecanemab trial—began to circulate online. Nearly half of the more than 200 scientists or medical practitioners who had signed the statement as of 20 December are recent consultants or grantees of one or both companies, Science has determined. (Some but not all disclosed a conflict of interest.)

The document describes lecanemab as a “foundational gamechanger” for the illness and calls for its approval and “no barrier” to widespread availability of the antibody. It notes possible safety concerns associated with ARIA but does not mention the deaths and serious brain injuries some have linked to the drug. FDA is expected to decide on lecanemab’s approval, and whether to require any warnings or cautions for prescribers, by 6 January 2023.

Smith, who has not signed the pro-lecanemab letter, acknowledges that people with early Alzheimer’s might deem the possibility of even very modest cognitive benefits worth the risk of debilitating cases of ARIA or even fatal outcomes. But he thinks any FDA approval should come with warnings.

An Alzheimer’s patient receiving lecanemab would require as many as five MRIs annually to properly monitor for ARIA, he says, and a vast education program would be needed to ensure that doctors outside of major medical centers can recognize problems found in the brain scans. Smith also calls for FDA to require a registry that records ARIA-related problems if it approves lecanemab.

Eisai’s spokesperson said that if lecanemab is approved, the company would work with FDA to ensure that doctors and patients “understand how to monitor the patient for side effects, such as ARIA,” adding that “for many patients the benefits will outweigh the risks.”

This story was supported by the Science Fund for Investigative Reporting.

Read original article here

USMNT knocked out of World Cup in round of 16 by clinical Netherlands



CNN
 — 

The United States Men’s National Team (USMNT) was defeated 3-1 by the Netherlands and knocked out of the World Cup in the round of 16, despite a spirited performance.

The Netherlands had been underwhelming during the group stages but showcased its quality and clinical edge on Saturday, with three well-taken goals from Memphis Depay, Daley Blind and Denzel Dumfries.

Haji Wright pulled one back for the USMNT late in the second half with his team trailing 2-1, but any comeback hopes were snuffed out by Dumfries’ 81st minute goal to all but confirm the Dutch victory.

US President Biden expressed pride in the USMNT’s performance in the World Cup following the team’s defeat , making a nod to the next World Cup in 2026, which will be held in the US as well as Canada and Mexico.

“Fellas, you made us proud. We get up and keep going,” Biden tweeted. “Here’s to a bright future and 2026 back here at home.”

The USMNT had more shots on goal than the Netherlands but once again, the lack of a proven goalscorer proved costly for Gregg Berhalter’s young team.

This was the USMNT’s youngest ever starting XI for a World Cup knockout match who lined up to face the Netherlands, aiming to reach the quarterfinals for just the second time in the modern era.

Berhalter’s side controlled the opening exchanges, finding space in behind the Dutch defense and Christian Pulisic had an early chance, going one-on-one with Oranje goalkeeper Andries Noppert who deflected the Chelsea forward’s shot away from goal.

Having soaked up that early pressure, the Netherlands then counterattacked with lethal precision.

After carving through the USMNT midfield, Cody Gakpo found Dumfries on the edge of the box who pulled the ball back to Depay perfectly positioned in front of the goal. The Barcelona forward finished off a sweeping move, firing the ball into the net.

The USMNT continued to press – a strike from Timothy Weah was batted away by Noppert and Sergiño Dest went on several mazy runs – but just could not find a way past the Dutch defense.

And then, on the stroke of halftime, the Netherlands doubled its lead as Blind rifled in Dumfries’ cross in an almost identical move to its first goal.

Immediately after the break, the USMNT almost pulled a goal back as the ball bobbled about in the box from a corner and Tim Ream guided it inches from the goal-line but the Netherlands cleared.

USMNT goalkeeper Matt Turner prrevent an almost certain goal as he made a double save, stopping two strikes in quick succession to keep his side’s fading hopes of reaching the quarterfinals alive.

His quick reflexes proved even more crucial minutes later when the USMNT finally scored as Wright latched onto Pulisic’s cross, with his effort looping over Noppert’s head.

The noise in the stadium, seemingly filled with American fans, briefly became deafening but just minutes later Dumfries hammered in Frenkie de Jong’s cross to confirm the Netherlands’ victory.

With that goal, Dumfries became the first Dutch player to be directly involved in more than three goals in a single World Cup match since 1978, according to the statistics website OptaJoe.

USMNT star Christian Pulisic said he was “really proud” of the team’s performances at the World Cup.

“I hope we showed a lot of people what we can do,” Pulisic told FOX’s Jenny Taft. “We deserved more from this tournament. End of the day, we couldn’t get it done, but I think there’s a lot we can be proud of.

“It hurts a lot right now,” added Pulisic. “We thought we could have advanced and done a lot more this time around, but I love these guys, and it’s a really special group that we have.”

The Netherlands now advances to the quarterfinals where it will face either Argentina or Australia on Friday.

Read original article here

HIV vaccine candidate induces immune response in early clinical trial



CNN
 — 

An experimental HIV vaccine has been found to induce broadly neutralizing antibodies among a small group of volunteers in a Phase 1 study. The findings suggest that a two-dose regimen of the vaccine, given eight weeks apart, can elicit immune responses against the human immunodeficiency virus.

The clinical trial results, published Thursday on World AIDS Day in the journal Science, establish “clinical proof of concept” in support of developing boosting regimens to induce immune responses against HIV infection, for which there is no cure and which can cause acquired immunodeficiency syndrome, known as AIDS.

The vaccine, called eOD-GT8 60mer, had a “favorable safety profile” and induced broadly neutralizing antibodies in 97%, or all but one, of the 36 recipients, according to the researchers from Scripps Research, the Fred Hutchinson Cancer Center, the National Institutes of Health and other institutions in the United States and Sweden.

Antibodies are proteins made by the immune system to help fight infections, and broadly neutralizing antibodies are known to neutralize many genetic variants of HIV, but they have been difficult to elicit by vaccination.

“Learning how to induce broadly neutralizing antibodies against pathogens with high antigenic diversity, such as HIV, influenza, hepatitis C virus, or the family of betacoronaviruses, represents a grand challenge for rational vaccine design,” the researchers wrote. “Germline-targeting vaccine design offers one potential strategy to meet this challenge.”

The eOD-GT8 60mer vaccine candidate is germline-targeting, meaning it was designed to induce the production of broadly neutralizing antibodies by targeting and stimulating the right antibody-producing cells.

The International AIDS Vaccine Initiative announced the start of this Phase 1 clinical trial in 2018, to evaluate the safety of eOD-GT8 60mer and the immune responses it is able to induce.

The trial included a total of 48 healthy adults, ages 18 to 50, who were enrolled at two sites: George Washington University in Washington and Fred Hutchinson Cancer Center in Seattle.

Among the participants, 18 received a 20-microgram dose of the vaccine and, eight weeks later, a same-size dose of the vaccine with an adjuvant; 18 received a 100-microgram dose of the vaccine and, eight weeks later, a same-size dose of the vaccine with an adjuvant; and 12 received two doses of a saline placebo, eight weeks apart. The adjuvant is called AS01B, developed by the pharmaceutical company GSK. The vaccines and placebo were given into the arm muscle.

The researchers collected and analyzed immune cells from the blood and lymph nodes of participants during the study. They specifically examined how B cells, a type of white blood cell that makes antibodies in the immune system, responded to the vaccine.

The researchers found no serious adverse events reported among the study participants, and no participants acquired HIV infection during the study. About 97% – or all but one – of the 48 study participants reported local or systemic adverse events that were generally mild or moderate, such as pain at the injection site, malaise and headache. In most cases, these events were resolved within a day or two.

After the first immunization, all vaccine recipients but no placebo recipients were found to produce antibodies elicited by the eOD-GT8 60mer vaccine. Those vaccine-induced responses increased after the second vaccination, the researchers wrote.

Another Phase 1 study on this vaccine candidate is underway, said Dr. Julie McElrath, senior vice president and director of the vaccine and infectious disease division at Fred Hutchinson Cancer Center, who was an author of the study.

What is unique about this HIV vaccine candidate is that it was engineered to directly target the production of broadly neutralizing antibodies, said Dr. Timothy Schacker, vice dean for research and program director in HIV medicine at the University of Minnesota Medical School, who was not involved in the research.

“In HIV, when we’ve designed and tested vaccines in the past, they didn’t for whatever reason induce these broadly neutralizing antibodies,” he said. “Call them super antibodies, if you want. The broadly neutralizing antibodies work more efficiently. They’re better at controlling things.”

By showing that broadly neutralizing antibodies can be induced by a vaccine, this new study could help inform the development of other types of immunizations, not just HIV vaccines, Schacker said.

“The hope is that if you can induce this kind of immunity in people, you can protect them from some of these viruses that we’ve had a very hard time designing vaccines for that are effective,” he said. “So this is an important step forward.”

Although this is “exciting science,” much more work needs to be done before this vaccine may be considered for use in the public, said Dr. Carlos del Rio, co-director of the Center for AIDS Research at Emory University and executive associate dean for Emory School of Medicine at Grady Health System, who was not involved in the new study.

“We know that broadly neutralizing antibodies are a potentially effective strategy to prevent HIV,” del Rio said. “We’re far from using this as a vaccine, but this is very exciting science. … Investing in this kind of research is critically important in not only developing a vaccine for HIV, but if this strategy works, it can be used for other vaccines.”

An HIV vaccine will probably need to elicit these broadly neutralizing antibodies, or bnAbs, “which are able to recognize globally diverse HIV strains and can prevent HIV infection. However, triggering bnAbs by vaccination has proven impossible so far. A key challenge is that bnAbs rarely develop, even during infection,” Penny Moore, of the University of the Witwatersrand and the National Institute for Communicable Diseases in South Africa, wrote in an editorial published alongside the new study.

A “key question” that still needs to be answered is how long the elicited antibodies from the first immunization can last.

Also, if the booster shot is too different from the previous vaccine, “antibodies that have been triggered by the first vaccination may not recognize the booster and will not mature further,” Moore wrote. “However, the incorporation of many different shots into an HIV vaccine regimen is unappealing. Getting the balance right between the need for antibody maturation toward bnAbs and feasibility in the real world will be essential.”

Last year, more than 38 million people were living with HIV or AIDS around the globe. More than 20 HIV vaccine clinical trials are ongoing around the world, according to the International AIDS Vaccine Initiative.

Many people in the United States have turned to daily HIV-prevention pills or frequent injections, known as PrEP, to reduce their risk of infection.

“It’s a daily pill or it’s a painful shot. It’s a shot that is uncomfortable at best that you have to get several times a year,” Schacker said of PrEP.

But having an HIV vaccine available would make protection against the virus more accessible, he said. “If you can give a vaccine, you’re going to reach more people and provide, if you have an effective vaccine, more and better coverage to reduce the probability of transmission if you’re exposed.”

Read original article here

Alzheimer’s disease: Experimental drug Lecanemab appears to slow progression in clinical trial but raises safety concerns



CNN
 — 

The experimental drug lecanemab shows “potential” as an Alzheimer’s disease treatment, according to new Phase 3 trial results, but the findings raise some safety concerns because of its association with certain serious adverse events.

Lecanemab has become one of the first experimental dementia drugs to appear to slow the progression of cognitive decline.

The long-awaited trial data, published Tuesday in the New England Journal of Medicine, comes about two months after drugmakers Biogen and Eisai announced that lecanemab had been found to reduce cognitive and functional decline by 27% in their Phase 3 trial.

A Phase 2 trial did not show a significant difference between lecanemab and a placebo in Alzheimer’s disease patients in 12 months – but the Phase 3 trial data suggests that at 18 months, lecanameb was associated with more clearance of amyloid and less cognitive decline.

“In persons with early Alzheimer’s disease, lecanemab reduced brain amyloid levels and was associated with less decline on clinical measures of cognition and function than placebo at 18 months but was associated with adverse events,” the researchers wrote. “Longer trials are warranted to determine the efficacy and safety of lecanemab in early Alzheimer’s disease.”

The Alzheimer’s Association said in a statement Tuesday that it welcomes and is further encouraged by the full Phase 3 data.

“These peer-reviewed, published results show lecanemab will provide patients more time to participate in daily life and live independently. It could mean many months more of recognizing their spouse, children and grandchildren. Treatments that deliver tangible benefits to those living with mild cognitive impairment (MCI) due to Alzheimer’s and early Alzheimer’s dementia are as valuable as treatments that extend the lives of those with other terminal diseases,” it says.

The Phase 3 trial was conducted at 235 sites in North America, Europe and Asia from March 2019 through March 2021. It involved 1,795 adults, ages 50 to 90, with mild cognitive impairment due to early Alzheimer’s disease or mild Alzheimer’s disease-related dementia.

About half of the participants were randomly assigned to receive lecanemab, given intravenously every two weeks, and the others received a placebo.

The researchers found that participants in both groups had a “clinical dementia rating” or CDR-SB score of about 3.2 at the start of the trial. Such a score is consistent with early Alzheimer’s disease, with a higher number associated with more cognitive impairment. By 18 months, the CDR-SB score went up 1.21 points in the lecanemab group, compared with 1.66 in the placebo group.

“Significant differences emerge as early as the six-month timepoint,” Dr. Christopher van Dyck, an author of the study and director of the Yale Alzheimer’s Disease Research Center, said Tuesday during a presentation at the Clinical Trials On Alzheimer’s Disease Conference in San Francisco.

“The lecanemab treatment met the primary and secondary endpoints,” he said.

Lecanemab, a monoclonal antibody, works by binding to amyloid beta, a hallmark of the degenerative brain disorder. At the start of the study, the participants’ average amyloid level was 77.92 centiloids in the lecanemab group and 75.03 centiloids in the placebo group.

By 18 months, the average amyloid level dropped 55.48 centiloids in the lecanemab group and went up 3.64 centiloids in the placebo group, the researchers found.

Based on these results, “lecanemab has the potential to make a clinically meaningful difference for people living with the early stages of Alzheimer’s disease and their families by slowing cognitive and functional decline,” Dr. Lynn Kramer, chief clinical officer of Alzheimer’s disease and brain health at Eisai, said in a news release.

About 6.9% of the trial participants in the lecanemab group discontinued the trial due to adverse events, compared with 2.9% of those in the placebo group. Overall, there were serious adverse events in 14% of the lecanemab group and 11.3% of the placebo group.

The most common adverse events in the drug group were reactions to the intravenous infusions and abnormalities on their MRIs, such as brain swelling and brain bleeding called amyloid-related imaging abnormalities, or ARIA.

“Lecanemab was generally well-tolerated. Most adverse events were infusion-related reactions, ARIA-H and ARIA-E and headache,” Dr. Marwan Sabbagh, an author of the study and professor at the Barrow Neurological Institute, said during Tuesday’s conference. He added that such events resolved within months.

ARIA brain bleeding was seen among 17.3% of those who received lecanemab and 9% of those in the placebo group; ARIA brain swelling was documented in 12.6% with lecanemab and 1.7% with placebo, according to the trial data.

Some people who get ARIA may not have symptoms, but it can occasionally lead to hospitalization or lasting impairment. And the frequency of ARIA appeared to be higher in people who had a gene called APOE4, which can increase the risk of Alzheimer’s disease and other dementias. ARIA “were numerically less common” among APOE4 noncarriers, the researchers wrote.

The researchers also wrote that about 0.7% of participants in the lecanemab group and 0.8% of those in the placebo group died, corresponding to six deaths documented in the lecanemab group and seven in the placebo group. “No deaths were considered by the investigators to be related to lecanemab or occurred with ARIA,” they wrote.

The company aims to file for approval of the drug in the United States by the end of March, according to its news release. The US Food and Drug Administration has granted lecanemab “priority review.”

In July, the FDA accepted Eisai’s Biologics License Application for lecanemab under the accelerated approval pathway, according to the company. The program allows for earlier approval of drugs that treat serious conditions and “fill an unmet medical need” while the drugs are being studied in larger and longer trials.

If the trials confirm that the drug provides a clinical benefit, the FDA grants traditional approval. But if the confirmatory trial does not show benefit, the FDA has regulatory procedures that could lead to taking the drug off the market.

“The FDA is expected to decide whether to grant accelerated approval to lecanemab by January 6, 2023,” the Alzheimer’s Association statement says. “Should the FDA do so, the current [Center for Medicare and Medicaid Services] policy will prevent thousands and thousands of Medicare beneficiaries with a terminal, progressive disease from accessing this treatment within the limited span of time they will have to access it. If a patient decides with their health care provider that a treatment is right for them, Medicare must stand behind them as it does for beneficiaries with every other disease.”

“If and when this drug is approved by the FDA, it will take clinicians some time to be able to parse out how this drug may or may not be effective in their own individual patients,” especially since carriers of the APOE4 gene could be at higher risk of side effects, said Dr. Richard Isaacson, adjunct associate professor of neurology at Weill Cornell Medicine, who is not involved in studying lecanemab or its development.

“While this study is certainly encouraging, how this translates to clinical practice, real-world clinical practice, remains to be seen,” he said of the Phase 3 trial data.

Overall, “physicians are starving for any possible therapy out there that can help our patients. I have four family members with Alzheimer’s disease. If I have a family member that comes to me and says, ‘Should I be on this drug?’ In the right patient, at the right dose, for the right duration, with adequate and careful monitoring for side effects, yes, I would suggest that this drug is a viable option,” Isaacson said. “I would say even an important option.”

He added that the experimental drug serves as an example of the important need for personalized medicine in the United States, especially when it comes to Alzheimer’s disease, such as using genetic testing in clinical practice to identify the APOE gene to better individualize the approach to a patient’s care.

“This is just the first chapter in what I hope to become a really long book in disease-modifying therapies for Alzheimer’s disease,” he said.

More than 300 Alzheimer’s treatments are in clinical trials, according to the Alzheimer’s Association.

Alzheimer’s disease was first documented in 1906, when Dr. Alois Alzheimer discovered changes in the brain tissue of a woman who had memory loss, language problems and unpredictable behaviors. The debilitating disease now affects more than 6 million adults in the United States.

There is no cure for Alzheimer’s disease, but there are several prescription drugs available to help manage symptoms. Last year, the FDA approved Aduhelm for early phases of Alzheimer’s disease. Before that, the FDA had not approved a novel therapy for the condition since 2003.

Although lecanemab is being tested as an Alzheimer’s drug, it is not a cure, said Tara Spires-Jones, deputy director of the Centre for Discovery Brain Sciences at the University of Edinburgh, who was not involved in the trial.

“Both groups in the trial had worsening symptoms, but people taking the drug did not decline as much in their cognitive skills,” Spires-Jones said in a written statement distributed by the UK-based Science Media Centre. “Longer trials will be needed to be sure that the benefits of this treatment outweigh the risks.”

In general, Alzheimer’s continues to be a “complex” disease, Bart De Strooper, director of the UK Dementia Research Institute, said in a statement distributed by the Science Media Centre.

“We still have a lot to learn about the underlying causes. It is therefore imperative that we continue to invest in discovery research, and through doing so, we may also identify new targets for which we can develop therapies we could use in combination with anti-amyloid drugs like lecanemab,” said De Strooper, who is a consultant for a series of pharmaceutical companies, including Eisai, but has not consulted on lecanemab.

“This trial proves that Alzheimer’s disease can be treated,” he said. “I hope we will start to see a reversal in the chronic underfunding of dementia research. I look forward to a future where we treat this and other neurodegenerative diseases with a battery of medications adapted to the individual needs of our patients.”

Read original article here

Lab-grown blood transfused to people in world-first clinical trial

Blood grown in a laboratory has been transfused into humans for the first time in a landmark clinical trial.

Future Publishing | Future Publishing | Getty Images

LONDON — Blood grown in a laboratory has been transfused into humans for the first time in a landmark clinical trial that U.K. researchers say could significantly improve treatment for people with blood disorders and rare blood types.

Two patients in the U.K. received tiny doses — equivalent to a few teaspoons — of the lab-grown blood in the first stage of a wider trial designed to see how it behaves inside the body.

The trial, which will now be extended to 10 patients over the course of several months, aims to study the lifespan of lab-grown cells compared with infusions of standard red blood cells.

Researchers say the aim is not to replace regular human blood donations, which will continue to make up the majority of transfusions. But the technology could allow scientists to manufacture very rare blood types which are difficult to source but which are vital for people who depend on regular blood transfusions for conditions such as sickle cell anemia.

“This world leading research lays the groundwork for the manufacture of red blood cells that can safely be used to transfuse people with disorders like sickle cell,” said Dr. Farrukh Shah, medical director of Transfusion for NHS Blood and Transplant, one of the collaborators on the project.

“The need for normal blood donations to provide the vast majority of blood will remain. But the potential for this work to benefit hard to transfuse patients is very significant,” she added.

How does the technology work?

The research, which was conducted by researchers in Bristol, Cambridge and London, as well as NHS Blood and Transplant, focuses on red blood cells that carry oxygen from the lungs to the rest of the body.

Initially, a regular donation of blood was taken and magnetic beads were used to detect flexible stem cells that are capable of becoming red blood cells.

Those stems were then placed in a nutrient solution in a laboratory. Over the course of around three weeks, the solution encouraged those cells to multiply and develop into more mature cells.

The cells were then purified using a standard filter — the same kind of filter that is used when regular blood donations are processed to remove white blood cells — before being stored and later transfused into the patients.

For the trial, the lab-grown blood was tagged with a radioactive substance, often used in medical procedures, to monitor how long it lasts in the body.

The same process will now be applied for a trial of 10 volunteers, who will each receive two donations of 5-10mls at least four months apart — one of normal blood and one of lab-grown blood — to compare the cells’ lifespans.

How much will it cost?

It is also hoped that a superior lifespan of lab-grown cells could mean patients require fewer transfusions over time.

A typical blood donation contains a mixture of young and old red blood cells, meaning their lifespan can be unpredictable and sub-optimal. Lab-grown blood, meanwhile, is freshly made, meaning it should last the 120 days expected of red blood cells.

Still, there are significant costs currently attached to the technology.

The average blood donation currently costs the NHS around £145, according to NHS Blood and Transplant. Lab-grown substitutes would likely be more expensive.

NHS Blood and Transplant said there was “no figure” for the procedure as yet, but added that costs would be reduced as the technology is scaled up.

“If the trial is successful and the research works, then it could be introduced at scale in future years, meaning that costs would fall,” a spokesperson told CNBC.

Read original article here

A man’s cancer vanished after he was injected with a weakened herpes virus in a promising clinical trial

FILE PHOTO: A patient undergoes a positron emission tomography scan.Photo by Vladimir SmirnovTASS via Getty Images

  • A new cancer therapy makes use of the herpes virus to fight harmful cells.

  • The modified virus infects cancer cells, causing them to explode, while alerting the immune system.

  • The therapy is in early trial stages but shows great promise, according to cancer researchers.

A new cancer therapy that uses a modified herpes virus to attack tumor cells showed promise in early clinical trials abroad.

The drug, called RP2, completely obliterated one patient’s oral cancer. The 39-year-old told the BBC that he had cancer of the salivary glands, which continued to grow despite attempts at treatment.

He was preparing for the end of his life when he learned about the experimental drug, which was available through a phase one safety trial at the Institute of Cancer Research in the UK.

After a short course of the drug, the patient — Krzysztof Wojkowski of west London — has been cancer-free for two years and counting, he told the BBC.

Other patients in the trial saw their tumors shrink, although the majority did not have a significant change: three out of nine patients who were given the trial drug alone, and seven of 30 who received a combined treatment, appeared to benefit from the experimental therapy.

While more research needs to be done to see how RP2 compares to known therapies, the drug seemed to help some patients and only caused mild side effects, such as tiredness. These early results are promising, said Jonathan Zager of the Moffitt Cancer Center, who was not involved in the trial.

“We’ll see some more studies done in the very near future, and I’m excited — certainly not disheartened or skeptical,” Zager told Insider.

A modified virus delivers a ‘one-two punch’ to cancer cells

The experimental therapy involves a weakened form of herpes simplex — the virus that causes cold sores — that has been modified to only infect tumors.

According to results presented at a medical conference in Paris, the viral therapy is engineered to selectively enter cancer cells while leaving normal cells alone. It’s injected directly into a tumor, while most other cancer drugs work systemically.

Once it has infiltrated, the virus replicates itself until the cancer cell explodes. What’s unique about RP2 is that it delivers a “one-two punch” against tumors, not only destroying the cells but rallying the immune system to attack what’s left, lead researcher Kevin Harrington said in a news release.

The drug works similarly to T-Vec, a viral therapy that was approved to treat advanced skin cancer in 2015. T-Vec was also engineered based on a herpes simplex virus and modified to include a gene that stimulates the production of immune cells, essentially preparing the immune system to attack.

These viral therapies hold great promise for treating multiple forms of cancer, with “truly impressive” treatment responses observed in patients with advanced esophageal cancer and a rare type of eye cancer, Harrington told the BBC.

The results are even more impressive considering that the patients recruited for clinical trials typically have tried several other treatments and surgeries to remove their cancers. Many, like Wojkowski, were out of options when they heard about RP2.

“When we have tumors that are heavily pretreated and they respond favorably — to RP2 or T-Vec — that’s even more food for thought, in the sense that now we have tumors that were resistant to treatment and are responding,” said Vager, who has treated hundreds of patients with T-Vec since it was approved.

According to Harrington, RP2 may work even better than T-Vec, if the early results are any indication.

“It’s had other modifications to the virus so that when it gets into cancer cells it effectively signs their death warrant,” Harrington told the BBC.

Read the original article on Insider

Read original article here

Lupus Pill Shows Promise in Mice; Clinical Trial Underway

Summary: Afimetoran, a newly developed pill to treat lupus, not only prevents lupus-like symptoms in mice, it also reverses signs of organ damage caused by the disease and prevents death. The medication is now undergoing phase 2 clinical trials to assess its effectiveness in lupus patients.

Source: American Chemical Society

Lupus is an autoimmune disease that attacks organs and can be fatal. There’s no cure, so current treatments aim to limit damage and ameliorate symptoms. Some of these therapies have to be injected, some have serious side effects, and many aren’t very effective.

But today, scientists report they have begun phase 2 clinical trials with a pill containing a compound that, in mice, not only prevents lupus-like symptoms, but also reverses signs of organ damage caused by the disease and prevents death.

The researchers will present their results at the fall meeting of the American Chemical Society (ACS). ACS Fall 2022 is a hybrid meeting being held virtually and in-person Aug. 21–25, with on-demand access available Aug. 26–Sept. 9. The meeting features nearly 11,000 presentations on a wide range of science topics.

“Few new therapies have succeeded, but we believe our compound could be an effective treatment for lupus,” says Alaric Dyckman, Ph.D. The disease affects 5 million people worldwide, according to the Lupus Foundation of America. Symptoms include rashes, extreme fatigue, pain, inflammation and deterioration of organs, such as the kidneys and heart, which can lead to death.

Lupus develops when the immune system attacks the body’s tissues. Years ago, researchers began suspecting that this process involved toll-like receptors (TLRs) 7 and 8, which are cellular proteins that activate the immune system when they detect viral RNA or mistakenly identify a person’s own RNA as a threat.

“Genetic data and evaluations of injectable treatments suggested TLR7 and 8 could be drug targets for lupus. What was missing was an ability to directly block these receptors with small molecules that could be taken orally,” says Dyckman. So in 2010, he and other scientists at Bristol Myers Squibb (BMS) set out to develop such compounds.

New options would be welcome, since many patients don’t respond fully to current medications. The two approved therapies that were specifically developed for lupus reduce activity of specific immune system components: AstraZeneca’s anifrolumab blocks a receptor for the protein interferon, while GlaxoSmithKline’s belimumab reduces the survival of white blood cells known as B cells.

Other treatments include steroids and other general immune suppressants, anti-malarials, anti-inflammatories and anticoagulants.

However, anifrolumab and belimumab must be given by injection or infusion, Dyckman notes, while steroids and general immune suppressants are associated with safety concerns and were not originally designed to treat lupus.

The BMS researchers began zeroing in on a suitable alternative by screening the company’s compound collection for molecules that could block TLR7/8 signaling. The team modified the structures of the initial hits to reduce interaction with other receptors, improve potency and enable oral dosing.

The resulting compound, “afimetoran,” binds to the target TLRs, inhibiting their operation to achieve beneficial activity. Like anifrolumab, it interferes with interferon, and like belimumab, it controls damage from overactive B cells. It also inhibits the production of multiple proinflammatory cytokines that cause a lot of tissue damage in lupus.

“With afimetoran, not only could we prevent the development of lupus-like symptoms in mice before their disease onset, but we could actually reverse the symptoms and prevent death in animals that were days or weeks away from succumbing to the disease,” Dyckman says.

“We hadn’t seen that reversal with other mechanisms we had evaluated, so we were particularly excited about that finding.”

The BMS researchers began zeroing in on a suitable alternative by screening the company’s compound collection for molecules that could block TLR7/8 signaling. Image is in the public domain

Dyckman says he believes the combined effects of afimetoran give it the potential to control lupus as well as or better than existing treatments and do so through an oral delivery, as opposed to requiring injection or infusion.

The team also found that afimetoran combined well with corticosteroid treatments in mice. That means patients might be able to use lower doses of steroids, a mainstay of lupus treatment.

Lower doses would be beneficial because steroids have side effects, such as weight gain, thinning bones, high blood pressure and diabetes, as well as an increased risk of infection.

See also

Phase 1 clinical trials of afimetoran to evaluate safety in healthy people and shed light on the compound’s behavior in the body have been completed.

The trials showed that a low, once-daily oral dose could almost completely block signaling through TLR7/8. And now, a phase 2 trial to test its effectiveness in lupus patients is underway. Because of its mode of action, Dyckman says, it may also work in other autoimmune disorders, such as psoriasis or arthritis.

BMS is testing other compounds against lupus, such as deucravacitinib, an oral, selective tyrosine kinase 2 (TYK2) inhibitor that is moving into phase 3 studies. Other companies are also making progress. Merck, for instance, is evaluating its own oral TLR7/8 blocker, enpatoran, in phase 2 trials.

But the crowded field doesn’t concern Dyckman. Despite intensive efforts to develop new therapies over the past several decades, few have succeeded.

“So getting a lot of shots on goal is important,” he says. “Also, lupus is such a heterogeneous disease that it’s unlikely that any single approach will provide relief for all of the patients out there.”

Funding: The researchers acknowledge support and funding from Bristol Myers Squibb.

About this neuropharmacology research news

Author: Katie Cottingham
Source: American Chemical Society
Contact: Katie Cottingham – American Chemical Society
Image: The image is in the public domain

Original Research: The findings will be presented at ACS Fall 2022

Read original article here

Lack of ‘high quality’ clinical guidelines for monkeypox may be hurting treatment efforts, researchers say  – The Hill

Story at a glance


  • A team of researchers mainly from the University of Oxford looked at 14 clinical guidelines for monkeypox and found that most of them were “low quality.” 

  • Researchers said the guidelines lacked sufficient detail on the virus and contradictory treatment recommendations.

  • The lack of standardization of these clinical guidelines could be hurting global efforts to treat people with the disease.  

A lack of up-to-date clinical guidelines on monkeypox may be impacting health care workers’ ability to administer effective and safe treatment in those infected, according to researchers.  

U.K.-based researchers looked at the existing clinical guidelines available for the disease and found that they often lack sufficient detail, are contradictory in their recommendations and fail to include impacts of the illness on different groups including children.  

The findings of the analysis were published earlier this week in the journal BMI Global Health.  


America is changing faster than ever! Add Changing America to your Facebook or Twitter feed to stay on top of the news.


Monkeypox is a viral disease that belongs to the same family as smallpox that can show up as a rash made up of small and painful blisters. Symptoms of the illness can also include fever, chills, fatigue, muscle aches, cough, neck pain and swollen lymph nodes.  

There has been an outbreak of the rare disease this summer with the U.S. Centers for Disease Control and Prevention confirming 14,115 cases of the virus across the country as of Thursday.  

Before the 2022 outbreak, the disease had been mainly reported in people living in about a dozen central and western African countries, according to the CDC.  

Now, a worldwide jump in cases has prompted the World Health Organization to declare the spike a public health emergency.  

In their analysis, researchers parsed through six major research databases with relevant information on monkeypox published up to October of last year and other “grey literature” like policy documents, newsletters and reports in multiple languages published until this May. They found a total of 14 relevant clinical guidelines, most of which were “low quality” and only covered a range of topics.  

Only five out of the 14 clinical guidelines gave any guidance on how to care for children infected with the virus and only three had any advice on how to treat pregnant women or people living with HIV who had contracted the disease, according to the analysis findings.  

Monkeypox treatment guidance was mostly limited to advice on antiviral medication and was not consistent, researchers added. Out of the guidelines reviewed, seven recommended that patients with monkeypox be given cidofovir, and out of those seven only four noted that the medication be only administered to treat severe infections.  

Only four clinical guidelines recommended tecovirimat and one advised giving the sick brincidofovir.  

Currently, the WHO recommends that health care professionals use tecovirimat to treat monkeypox patients instead of cidofovir.  

None of the clinical guidelines reviewed contained details on optimal doses, timing or length of treatment and only one had any recommendations on supportive care and treatment complications, according to the findings.  

Every one of the 14 guidelines encouraged vaccination as post-exposure prophylaxis but not all of them had up to date information on the newer generation of vaccines.  

Guidelines are key tools for clinicians and are especially important for emerging infectious diseases that health care workers might be less familiar with, according to Louise Sigfrid, who works at the ISARIC Global Support Center, Pandemic Sciences Institute at the University of Oxford and is one of the lead researchers in the analysis.  

Sigfrid noted that some of her colleagues in London that treated monkeypox patients early in the U.K.’s outbreak this spring reported struggling to know how to diagnose and treat patients due to limited guidelines.  

“The main aim of the clinical national guidelines is to benefit patient care,” Sigfrid told Changing America. “It’s also to standardize care. To make sure that all patients in all settings have access to evidence-based care and that clinicians have access to evidence-based treatments.”  

Standardizing clinical guidelines also serves an important purpose when it comes to developing vaccines for diseases like monkeypox, Sigfrid added. Without them, clinical research for treatments can be weakened and hamper randomized control trials.  

“As rare diseases are emerging…what we have also seen when we are trying to implement rapidly is that clinicians in different sites will use different treatments,” said Sigfrid.  


READ MORE STORIES FROM CHANGING AMERICA

BLIND WOMAN CAN SEE SHAPES WITH HELP FROM NEW BRAIN IMPLANTS

RATS RUNNING THROUGH VR MAZE OFFER INSIGHTS ON MEMORY AND LEARNING

AUTO-INJECTING PILL COULD DELIVER DRUGS NORMALLY GIVEN BY SHOTS

IN A BREAKTHROUGH CARTILAGE FROM NOSE IS USED TO TREAT SEVERE KNEE PROBLEMS

Published on Aug. 19, 2022



Read original article here

Clinical Management Guidelines Lacking for Monkeypox | Madison.com Health, Sports Health & Fitness

FRIDAY, Aug. 19, 2022 (HealthDay News) — There is a lack of evidence-based clinical management guidelines for monkeypox (MPX), according to a review published online Aug. 16 in BMJ Global Health.

Eika Webb, M.B.B.S., from Liverpool School of Tropical Medicine in the United Kingdom, and colleagues conducted a systematic review and identified MPX guidelines providing treatment and supportive care recommendations. A total of 14 guidelines were included, from 2,026 records screened.

The researchers found that most guidelines were of low quality (median score, 2 out of 7), lacked detail, and covered only a narrow range of topics. Most of the guidelines focused on adults; five, three, and three provided some advice for children, pregnant women, and people living with HIV, respectively. Treatment guidance was mainly limited to antiviral advice; cidofovir was advised in seven guidelines (four for severe MPX only), while four and one guidelines advised tecovirimat and brincidofovir, respectively. Recommendations on supportive care and treatment of complications were provided in only one guideline. Vaccination was recommended as postexposure prophylaxis (PEP) in all guidelines. Vaccinia immune globulin was advised as PEP for severe cases in people with immunosuppression in three guidelines.

“Urgent investments into research to identify optimal treatment and prophylaxis strategies are needed for the whole population, in any setting, to benefit patient care and outcomes,” the authors write.

Read original article here

Clinical Trial Restored Sight to 20 People With Corneas Made From An Unlikely Source : ScienceAlert

Implants made from pig skin have restored sight to 20 people with diseased corneas, in an exciting pilot clinical trial. Many of the patients were blind before receiving the help of this bioengineered tissue.

Incredibly, after two years, all 14 of those blind people had their vision restored and three of them, and three of them now have perfect 20/20 vision.

“This gets us around the problem of [a] shortage of donated corneal tissue and access to other treatments for eye diseases,” says Linköping University ophthalmology researcher Neil Lagali.

While around 12.7 million people suffer vision loss due to problems with their corneas, only 1 in 70 manage to receive a cornea transplant – the only way to restore their vision.

As the means to provide these transplants are costly, and donated corneas are in short supply, most people in the world do not have access to effective treatments.

“We’ve made significant efforts to ensure that our invention will be widely available and affordable by all and not just by the wealthy. That’s why this technology can be used in all parts of the world,” says Linköping University biomedical engineer Mehrdad Rafat.

To achieve this, Rafat and colleagues developed a new technique that requires no stitches so doctors can perform the implant procedure with less specialized conditions and equipment.

“A less invasive method could be used in more hospitals, thereby helping more people. With our method, the surgeon doesn’t need to remove the patient’s own tissue. Instead, a small incision is made, through which the implant is inserted into the existing cornea,” explains Lagali.

What’s more, the material used to create the implant is a byproduct of the food industry and, thanks to specially developed packaging and sterilization processes, the final product can be stored for up to two years. In contrast, donated human corneas must be used within two weeks.

Our cornea – the clear screen over the front part of our eye that shields our iris and pupil – is mostly composed of different types of collagen. This structure can gradually thin out over time, causing it to bulge outwards and distort our vision in a condition called keratoconus.

While the exact cause of this thinning isn’t known, genetics, vigorous eye rubbing, and conditions including hay fever, asthma, Down syndrome, and Ehlers-Danlos syndrome can increase the chances of developing keratoconus.

So the researchers purified collagen from pig skin to create a new cornea layer. They used chemical and photochemical methods to strengthen this usually soft material, making it more stable, resulting in a hydrogel they’ve called a bioengineered porcine construct, double crosslinked (BPCDX).

Changes in corneal thickness with arrows indicating the implant outline post operation (bottom). (Rafat et al, Nature Biotechnology, 2022)

Refining their techniques in animal models, researchers then developed a simple method to insert BPCDX into the recipient’s cornea, eliminating the need to remove the existing tissue.

Here, the implant flattens the cornea’s buckling and provides the lost thickness, repairing the eye’s capacity to focus.

The minimally invasive surgery leaves the corneal nerves and cell layers intact, allowing the wound to rapidly heal.

Following implantation through a 2-millimeter incision, the BPCDX successfully remained transparent. There was no scar formation or adverse reaction, and no intensive therapy or further surgery was required; just an eight-week treatment with immunosuppressive eye drops and a bandage.

The bioengineered cornea checked all the safety boxes.

After two years, the participants from Iran and India experienced an average increase of more than 200 micrometers in their cornea’s thickness, and a decrease in its curvature, improving their vision at least to the extent of traditional corneal transplants.

Previously attempted biomaterial implants into the eye ended up thinning, but the fortified pig cell collagen held strong and kept the implant stable, even after eight years, the team reported based on their previous studies and unpublished data.

“No previous study has, to our knowledge, achieved full corneal transparency in vivo with sufficient corneal thickening and flattening, or with significant visual acuity gains as reported here,” the researchers wrote in their paper.

A larger clinical trial is now being planned, but if the pilot is any indication the researchers are hopeful for additional promising results that will help the new procedure meet regulatory approvals.

“The results show that it is possible to develop a biomaterial that meets all the criteria for being used as human implants, which can be mass-produced and stored up to two years and thereby reach even more people with vision problems,” Lagali concludes.

This research was published in Nature Biotechnology.

Read original article here