User login
IUD injury risk rises shortly after women give birth
Women for whom an intrauterine device is inserted from 4 days to 6 weeks after giving birth, as well as those who are breastfeeding, are at higher risk of the contraceptive device puncturing their uterus, new research shows.
The risk of perforation was nearly seven times higher for patients who received an IUD within that window than for those with an IUD who’d never given birth or who were more than a year out from delivery, the researchers found. Health care providers should make patients aware of the heightened risk and should monitor these patients more closely, according to Susan Reed, MD, an ob.gyn. at the University of Washington, Seattle, lead author of the new study.
“I’m a surgeon, and I like to be able to give people good information and good data about risks and benefits for their choices,” Dr. Reed told this news organization. “Uterine perforations related to IUDs are exceedingly rare, and to get good data or known risk factors, you need huge studies. This was the largest study done that really provided accurate information for patients and providers.” The new study, which appears in a recent issue of The Lancet, also found that the risk of uterine perforation was lower if an IUD had been inserted immediately after delivery.
Dr. Reed and colleagues analyzed data from the health records of 326,658 women younger than 50 years for whom an IUD was inserted between 2001 and 2018 at four health care systems. Nearly 30% of these patients received an IUD after giving birth.
The researchers identified a total of 1,008 uterine perforations, for a cumulative incidence at 5 years of 0.6%. The cumulative incidence of uterine perforations was lowest in the group of women who were considered “nonpostpartum”; these women either received an IUD a full year after giving birth or had not given birth during the study period (0.29%; 95% confidence interval, 0.26-0.34).
Women who received an IUD during the 3 days after delivery had a nearly threefold increased risk of an IUD perforation over nonpostpartum women.
In addition, the cumulative incidence of perforation was almost double among breastfeeding women, compared with women who were not breastfeeding. However, Dr. Reed and coauthors noted that breastfeeding is highly beneficial for babies and that the risk of IUD perforation is relatively small.
Among the women who received an IUD following birth, Dr. Reed’s group found that 673 uterine perforations – of which 62% were complete – occurred in breastfeeding individuals, 37% more than for those who did not breastfeed.
Dr. Reed said the study provided some clarity on previous notions that women who’d never given birth were possibly at higher risk for uterine perforation because of smaller uteruses.
“We used to be concerned that women who had never had a pregnancy at all might be at higher risk because their uterus was smaller, the cervix was tighter, and therefore perhaps they might have greater risks,” Dr. Reed said in an interview. “As a clinician and as a provider, it’s pretty exciting to me to be able to tell our younger women who have never had a pregnancy that indeed their risk is lower than anybody else’s.”
The findings help women to make informed decisions, but overall, the benefits of IUDs outweigh the risks, said Monica V. Dragoman, MD, assistant professor of obstetrics, gynecology, and reproductive science at the Icahn School of Medicine at Mount Sinai, New York.
“The likelihood of anyone experiencing these types of complications of that population level remains really low,” Dr. Dragoman said.
The findings also provide guidance for providers as to which patients should come in for additional follow-up visits following insertion, Dr. Reed said.
“These are small risks, but it does tell us where we need to consider if there’s a challenging insertion,” Dr. Reed said. “You’re going to look with the ultrasound and make sure the placement looks right. You’re going to give instructions that if the woman has pain or a change in her bleeding pattern, you want to see her back.”
Patients should be aware of the symptoms of uterine perforation – an abrupt change in bleeding pattern and pelvic pain. Perforation correction typically consists of a minimally invasive surgical procedure.
The study was conducted as a result of an order from the Food and Drug Administration to Bayer Pharmaceuticals to evaluate risks of uterine perforation for women who’d received the company’s Mirena IUD. The findings led the company to update the language on the packaging of the device so as to specify the main symptoms of uterine perforations.
The study authors received research funding from Bayer. Multiple authors are employees of Bayer. One study author has in the past received funding from CooperSurgical, Bayer Healthcare Pharmaceutical, and Merck. Bayer was provided the opportunity to review the manuscript before submission, and comments were advisory only.
A version of this article first appeared on Medscape.com.
Women for whom an intrauterine device is inserted from 4 days to 6 weeks after giving birth, as well as those who are breastfeeding, are at higher risk of the contraceptive device puncturing their uterus, new research shows.
The risk of perforation was nearly seven times higher for patients who received an IUD within that window than for those with an IUD who’d never given birth or who were more than a year out from delivery, the researchers found. Health care providers should make patients aware of the heightened risk and should monitor these patients more closely, according to Susan Reed, MD, an ob.gyn. at the University of Washington, Seattle, lead author of the new study.
“I’m a surgeon, and I like to be able to give people good information and good data about risks and benefits for their choices,” Dr. Reed told this news organization. “Uterine perforations related to IUDs are exceedingly rare, and to get good data or known risk factors, you need huge studies. This was the largest study done that really provided accurate information for patients and providers.” The new study, which appears in a recent issue of The Lancet, also found that the risk of uterine perforation was lower if an IUD had been inserted immediately after delivery.
Dr. Reed and colleagues analyzed data from the health records of 326,658 women younger than 50 years for whom an IUD was inserted between 2001 and 2018 at four health care systems. Nearly 30% of these patients received an IUD after giving birth.
The researchers identified a total of 1,008 uterine perforations, for a cumulative incidence at 5 years of 0.6%. The cumulative incidence of uterine perforations was lowest in the group of women who were considered “nonpostpartum”; these women either received an IUD a full year after giving birth or had not given birth during the study period (0.29%; 95% confidence interval, 0.26-0.34).
Women who received an IUD during the 3 days after delivery had a nearly threefold increased risk of an IUD perforation over nonpostpartum women.
In addition, the cumulative incidence of perforation was almost double among breastfeeding women, compared with women who were not breastfeeding. However, Dr. Reed and coauthors noted that breastfeeding is highly beneficial for babies and that the risk of IUD perforation is relatively small.
Among the women who received an IUD following birth, Dr. Reed’s group found that 673 uterine perforations – of which 62% were complete – occurred in breastfeeding individuals, 37% more than for those who did not breastfeed.
Dr. Reed said the study provided some clarity on previous notions that women who’d never given birth were possibly at higher risk for uterine perforation because of smaller uteruses.
“We used to be concerned that women who had never had a pregnancy at all might be at higher risk because their uterus was smaller, the cervix was tighter, and therefore perhaps they might have greater risks,” Dr. Reed said in an interview. “As a clinician and as a provider, it’s pretty exciting to me to be able to tell our younger women who have never had a pregnancy that indeed their risk is lower than anybody else’s.”
The findings help women to make informed decisions, but overall, the benefits of IUDs outweigh the risks, said Monica V. Dragoman, MD, assistant professor of obstetrics, gynecology, and reproductive science at the Icahn School of Medicine at Mount Sinai, New York.
“The likelihood of anyone experiencing these types of complications of that population level remains really low,” Dr. Dragoman said.
The findings also provide guidance for providers as to which patients should come in for additional follow-up visits following insertion, Dr. Reed said.
“These are small risks, but it does tell us where we need to consider if there’s a challenging insertion,” Dr. Reed said. “You’re going to look with the ultrasound and make sure the placement looks right. You’re going to give instructions that if the woman has pain or a change in her bleeding pattern, you want to see her back.”
Patients should be aware of the symptoms of uterine perforation – an abrupt change in bleeding pattern and pelvic pain. Perforation correction typically consists of a minimally invasive surgical procedure.
The study was conducted as a result of an order from the Food and Drug Administration to Bayer Pharmaceuticals to evaluate risks of uterine perforation for women who’d received the company’s Mirena IUD. The findings led the company to update the language on the packaging of the device so as to specify the main symptoms of uterine perforations.
The study authors received research funding from Bayer. Multiple authors are employees of Bayer. One study author has in the past received funding from CooperSurgical, Bayer Healthcare Pharmaceutical, and Merck. Bayer was provided the opportunity to review the manuscript before submission, and comments were advisory only.
A version of this article first appeared on Medscape.com.
Women for whom an intrauterine device is inserted from 4 days to 6 weeks after giving birth, as well as those who are breastfeeding, are at higher risk of the contraceptive device puncturing their uterus, new research shows.
The risk of perforation was nearly seven times higher for patients who received an IUD within that window than for those with an IUD who’d never given birth or who were more than a year out from delivery, the researchers found. Health care providers should make patients aware of the heightened risk and should monitor these patients more closely, according to Susan Reed, MD, an ob.gyn. at the University of Washington, Seattle, lead author of the new study.
“I’m a surgeon, and I like to be able to give people good information and good data about risks and benefits for their choices,” Dr. Reed told this news organization. “Uterine perforations related to IUDs are exceedingly rare, and to get good data or known risk factors, you need huge studies. This was the largest study done that really provided accurate information for patients and providers.” The new study, which appears in a recent issue of The Lancet, also found that the risk of uterine perforation was lower if an IUD had been inserted immediately after delivery.
Dr. Reed and colleagues analyzed data from the health records of 326,658 women younger than 50 years for whom an IUD was inserted between 2001 and 2018 at four health care systems. Nearly 30% of these patients received an IUD after giving birth.
The researchers identified a total of 1,008 uterine perforations, for a cumulative incidence at 5 years of 0.6%. The cumulative incidence of uterine perforations was lowest in the group of women who were considered “nonpostpartum”; these women either received an IUD a full year after giving birth or had not given birth during the study period (0.29%; 95% confidence interval, 0.26-0.34).
Women who received an IUD during the 3 days after delivery had a nearly threefold increased risk of an IUD perforation over nonpostpartum women.
In addition, the cumulative incidence of perforation was almost double among breastfeeding women, compared with women who were not breastfeeding. However, Dr. Reed and coauthors noted that breastfeeding is highly beneficial for babies and that the risk of IUD perforation is relatively small.
Among the women who received an IUD following birth, Dr. Reed’s group found that 673 uterine perforations – of which 62% were complete – occurred in breastfeeding individuals, 37% more than for those who did not breastfeed.
Dr. Reed said the study provided some clarity on previous notions that women who’d never given birth were possibly at higher risk for uterine perforation because of smaller uteruses.
“We used to be concerned that women who had never had a pregnancy at all might be at higher risk because their uterus was smaller, the cervix was tighter, and therefore perhaps they might have greater risks,” Dr. Reed said in an interview. “As a clinician and as a provider, it’s pretty exciting to me to be able to tell our younger women who have never had a pregnancy that indeed their risk is lower than anybody else’s.”
The findings help women to make informed decisions, but overall, the benefits of IUDs outweigh the risks, said Monica V. Dragoman, MD, assistant professor of obstetrics, gynecology, and reproductive science at the Icahn School of Medicine at Mount Sinai, New York.
“The likelihood of anyone experiencing these types of complications of that population level remains really low,” Dr. Dragoman said.
The findings also provide guidance for providers as to which patients should come in for additional follow-up visits following insertion, Dr. Reed said.
“These are small risks, but it does tell us where we need to consider if there’s a challenging insertion,” Dr. Reed said. “You’re going to look with the ultrasound and make sure the placement looks right. You’re going to give instructions that if the woman has pain or a change in her bleeding pattern, you want to see her back.”
Patients should be aware of the symptoms of uterine perforation – an abrupt change in bleeding pattern and pelvic pain. Perforation correction typically consists of a minimally invasive surgical procedure.
The study was conducted as a result of an order from the Food and Drug Administration to Bayer Pharmaceuticals to evaluate risks of uterine perforation for women who’d received the company’s Mirena IUD. The findings led the company to update the language on the packaging of the device so as to specify the main symptoms of uterine perforations.
The study authors received research funding from Bayer. Multiple authors are employees of Bayer. One study author has in the past received funding from CooperSurgical, Bayer Healthcare Pharmaceutical, and Merck. Bayer was provided the opportunity to review the manuscript before submission, and comments were advisory only.
A version of this article first appeared on Medscape.com.
FROM THE LANCET
$3 billion in cancer drug waste: Can it be salvaged?
Three billion dollars: It’s enough to finance the annual out-of-pocket costs for 1 in 7 patients with cancer. It would cover almost half of the National Cancer Institute’s annual budget. And it could fund President Biden’s entire Cancer Moonshot program, with more than a billion to spare.
It’s also how much the United States spends on unused cancer drugs each year, some experts estimate.
Drug companies typically sell infused drugs in one or two single-dose vial sizes, but patients don’t come in such neat packages. A patient may need 300 mg of a drug that is only sold as 200 mg vials, which means half of a vial will go to waste.
Although most oncology drugs don’t incur substantial waste, even small volumes can translate to millions of dollars a year.
But can this money be saved or reallocated, if only we delivered drugs more efficiently?
Some experts don’t believe that’s possible.
“Attempts to recoup money for discarded drugs wouldn’t happen in a vacuum,” said Robin Yabroff, PhD, MBA, an epidemiologist and scientific vice president of Health Services Research at the American Cancer Society, who was part of a committee commissioned to evaluate the costs associated with discarded drugs.
The potential catch of any widespread effort to seek repayment or reduce the amount of discarded drugs, Dr. Yabroff and colleagues note, is that manufacturers would “simply increase the price of the vial.”
In other words, attempting to fix one problem may lead to another — essentially a whack-a-mole of cancer costs, which are projected to balloon to $246 billion by 2030.
What this means is without sweeping policies to rein in cancer care costs, oncologists can only do so much. And every little bit counts.
“We are left chipping away at this monster of cancer care costs,” said Adam Binder, MD, a medical oncologist at the Sidney Kimmel Cancer Center at Jefferson Health in Philadelphia.
Millions spent on “reasonable amount” of waste
Michal Sarfaty, MD, was excited when enfortumab vedotin came on the market to treat advanced urothelial cancer in late 2019.
The cost of the drug, however, tempered her enthusiasm.
Enfortumab vedotin is a “great drug,” said Dr. Sarfaty, an oncologist at the Sheba Medical Center, Ramat Gan, Israel. But it can cost upwards of $500,000 a year for an average-weight man.
Given the expense, Dr. Sarfaty wanted to understand how much of the drug gets thrown away. During a fellowship at Memorial Sloan Kettering (MSK) Cancer Center in New York, Dr. Sarfaty explored the amount of unused enfortumab vedotin among the 64 patients who received the drug in 2020. She, along with a team at MSK, calculated the price tag of that waste and extrapolated those estimates for patients across the country.
Although waste occurred in almost half of administered doses (367 of 793), only a small volume got discarded — 2.9% per dose, on average.
Multiplying unused milligrams by the cost per milligram, Dr. Sarfaty and colleagues estimated that, for each patient, $3,127 of the drug got discarded. When calculated over the year, the cost came to just over $200,000 at MSK, and nearly $15 million when projected across the approved patient population in the United States.
“Ultimately, we did not see a lot of waste with this specific drug,” Dr. Sarfaty said. “Under 2.9% is considered a reasonable amount, below the 3% threshold Peter Bach, MD, and colleagues recommend. But even with this small amount of waste, the cost per patient and to the system remains notable.”
The problem with recouping drug waste
Estimates from the Centers for Medicare & Medicaid Services (CMS), which tracks costs associated with discarded weight-based drugs covered under Medicare Part B, support the notion that small quantities of discarded drugs can still translate to big bucks.
Since 2017, CMS has required healthcare providers to report the volume of drugs discarded from a single-dose vial using a code, known as the JW modifier. The JW modifier means that providers can be reimbursed for the entire vial amount, not just the quantity the patient used.
In 2019, claims data from Medicare Part B showed that 1.85% of discarded rituximab came to $33.3 million. For infliximab, the 1.55% of discarded liquid translated to $15 million, and just 0.36% of discarded pembrolizumab reached $10 million.
However, experts question whether the JW modifier accurately reflects the quantity of drugs discarded.
According to the 2021 report from the National Academies of Sciences, Engineering, and Medicine (NASEM), most physicians don’t use the JW modifier. Among Medicare claims, 16.2% included the JW modifier in 2017 and 16.9% did in 2018.
The rate was significantly lower for private insurance. Of more than 4 million private insurance claims on 77 drugs made in 2017 and 2018, only 3.6% included the JW modifier; 15 of these drugs had no JW claims.
“Although we found that most physicians don’t use the JW modifier, even those who do, don’t use it consistently, even for the same patient,” said Dr. Yabroff, a co-author on the report.
Going a step further, Dr. Yabroff and colleagues argue that even if everyone used the JW modifier as intended, manufacturers would probably increase the price of drugs to compensate for any loss, potentially eliminating savings for payers.
That’s because, in the United States, manufacturers typically base drug prices on a patient and payers’ “willingness to pay for better health,” not on the volume of liquid used. Take a patient who pays $2,000 to receive the dose they need. If that dose is 600 mg but requires using two vials of 400 mg, then “to the patient, the 600-mg dose is worth $2,000, and the remainder has no value whatsoever,” the NASEM authors argue.
The authors parallel this scenario to purchasing a designer coat or dress. If that item requires alterations that remove a section of material, “the customer does not typically get a rebate because all the fabric was not needed,” the NASEM team writes.
But there’s a flaw in this rationale, argues Daniel Goldstein, MD, a medical oncologist at the Rabin Medical Center, Petah Tikva, Israel. A person’s willingness to pay for better health assumes that the price of a drug is based on proper market forces, where a drug’s cost and its effectiveness are in harmony.
“The problem is we’re operating in a broken market where the prices of oncology drugs have no real bearing on their efficacy,” said Dr. Goldstein.
And, as Dr. Bach noted in a 2021 Health Affairs piece, willingness to pay also requires that consumers know what they’re paying and allows them to walk away from an excessively high price.
But neither is a reality.
For one, Dr. Bach explains, companies may lowball the monthly price of a drug. In 2020, GlaxoSmithKline (GSK) announced that its new drug Blenrep would carry a list price of $8,277 per vial, or about $23,900 per month for an average 79 kg (175 lb) patient. That price accounts for two vials of the drug. But, according to Dr. Bach, “what GSK left out is that 44% of U.S. adults weigh more than 80 kg, and above that weight, three vials are needed per dose.” That would raise the average monthly cost to $30,479.
Perhaps more importantly, consumers can’t easily walk away.
“Medicare can’t negotiate prices and is forced to pay what a drug company says,” Dr. Goldstein said. “This is very different to when I buy a coat. If the price is too high, I can walk away.”
Fixed dosing: A solution or a new problem?
Efforts to reduce the financial impact of discarded cancer drugs can blow back on physicians, patients, and payers in other unanticipated ways. Take fixed dosing. Although chemotherapy dosing remains weight-based, many targeted therapies — such as nivolumab and pembrolizumab — recently transitioned to a fixed dosing regimen.
Administering a fixed, instead of weight-based, dose eliminates waste but can create new problems.
“Patients with cancer not only tend to get too high a dose of the drug, but costs go up significantly,” said Dr. Goldstein. In a 2017 analysis, Dr. Goldstein and colleagues compared dosing strategies in patients with metastatic non–small cell lung cancer who received pembrolizumab. The team found that the total annual cost of weight-based dosing was $2.6 billion, whereas the cost of the fixed dosing strategy was $3.44 billion — 24% more. In other words, personalized weight-based dosing would save more than $825 million dollars in the United States each year.
A 2020 analysis based in France found a similar cost increase of 26% for fixed dosing of pembrolizumab as well as nivolumab.
“I’ve argued we should go back to weight-based dosing,” Dr. Goldstein said. “Why should we give a higher dose with the same efficacy when that dose will cost significantly more and has the potential to increase adverse events?”
Does dose rounding work?
Rose DiMarco, PharmD, BCPS, BCOP, keeps a tight watch on patients being treated at the Sidney Kimmel Cancer Center at Jefferson Health in Philadelphia.
Dr. DiMarco educates patients about their treatment plan, reviews their lab results, and monitors them for side effects and drug interactions.
She also thinks a lot about costs.
“We spend about $100,000 a day on oncology drugs, and we want to make sure we’re not being wasteful,” Dr. DiMarco said in an interview.
One major initiative to curb waste and reduce costs at Jefferson has centered on dose rounding, which calculates whether a specific dose can be altered slightly to conserve vials and prevent waste. According to the Hematology/Oncology Pharmacy Association, a patient can receive up to 10% more or less of a weight-based dose without impacting treatment efficacy.
If, for instance, a patient requires 380 mg, but two vials come to 400 mg, rounding up that dose by approximately 5% means eliminating 20 mg that would go unused. But if that patient requires 420 mg, rounding down about 5% means substantial savings from not opening a new vial.
At Jefferson, Dr. DiMarco and her pharmacy colleagues map out dose ranges for all patients. Anyone who falls inside the 10% may be eligible for dose rounding. Anyone who doesn’t will receive the usual dose.
Although it is a challenge to implement, dose rounding has become standard of care at many cancer centers across the United States and is linked to substantial savings.
A 2018 analysis projected annual savings of $865,000 associated with rounding down eight monoclonal antibodies for patients with metastatic disease at a community cancer center. A more recent analysis from the Mayo Clinic found that dose rounding saved a total of 9,814 drug vials — 4485 of which were cancer drugs and 5329 of which were biologics — and resulted in $7.3 million in savings over 6 months in 2019 — $1.56 million from oncology agents and $5.7 from biologics.
And in a small 2019 analysis, researchers at Jefferson showed dose rounding of one monoclonal antibody saved approximately $30,000 in just 3 months, Dr. DiMarco noted.
“Not only does this process reduce costs and waste, but it also standardizes the preparation of hazardous medications, which can help prevent medication errors,” Dr. DiMarco said.
Nibbling around the edges
Despite estimates that scale into the billions of dollars, “drug wastage is just a small part of overall cancer costs,” Dr. Sarfaty said.
Fumiko Chino, MD, a radiation oncologist at MSK, agrees. “When we talk about affordability and cost, we can nibble around the edges of what’s really important,” Dr. Chino said. “Discarded drugs may cost a lot when you consider them in aggregate, but they are not as important as negotiated drug prices, which could substantially reduce overall costs.”
And until drug prices are addressed on a broader policy level, the cost of cancer care likely won’t improve in a meaningful way.
“But for the patient sitting in front of me, my focus will always be to provide the best care possible,” Dr. Binder said.
A version of this article first appeared on Medscape.com.
Three billion dollars: It’s enough to finance the annual out-of-pocket costs for 1 in 7 patients with cancer. It would cover almost half of the National Cancer Institute’s annual budget. And it could fund President Biden’s entire Cancer Moonshot program, with more than a billion to spare.
It’s also how much the United States spends on unused cancer drugs each year, some experts estimate.
Drug companies typically sell infused drugs in one or two single-dose vial sizes, but patients don’t come in such neat packages. A patient may need 300 mg of a drug that is only sold as 200 mg vials, which means half of a vial will go to waste.
Although most oncology drugs don’t incur substantial waste, even small volumes can translate to millions of dollars a year.
But can this money be saved or reallocated, if only we delivered drugs more efficiently?
Some experts don’t believe that’s possible.
“Attempts to recoup money for discarded drugs wouldn’t happen in a vacuum,” said Robin Yabroff, PhD, MBA, an epidemiologist and scientific vice president of Health Services Research at the American Cancer Society, who was part of a committee commissioned to evaluate the costs associated with discarded drugs.
The potential catch of any widespread effort to seek repayment or reduce the amount of discarded drugs, Dr. Yabroff and colleagues note, is that manufacturers would “simply increase the price of the vial.”
In other words, attempting to fix one problem may lead to another — essentially a whack-a-mole of cancer costs, which are projected to balloon to $246 billion by 2030.
What this means is without sweeping policies to rein in cancer care costs, oncologists can only do so much. And every little bit counts.
“We are left chipping away at this monster of cancer care costs,” said Adam Binder, MD, a medical oncologist at the Sidney Kimmel Cancer Center at Jefferson Health in Philadelphia.
Millions spent on “reasonable amount” of waste
Michal Sarfaty, MD, was excited when enfortumab vedotin came on the market to treat advanced urothelial cancer in late 2019.
The cost of the drug, however, tempered her enthusiasm.
Enfortumab vedotin is a “great drug,” said Dr. Sarfaty, an oncologist at the Sheba Medical Center, Ramat Gan, Israel. But it can cost upwards of $500,000 a year for an average-weight man.
Given the expense, Dr. Sarfaty wanted to understand how much of the drug gets thrown away. During a fellowship at Memorial Sloan Kettering (MSK) Cancer Center in New York, Dr. Sarfaty explored the amount of unused enfortumab vedotin among the 64 patients who received the drug in 2020. She, along with a team at MSK, calculated the price tag of that waste and extrapolated those estimates for patients across the country.
Although waste occurred in almost half of administered doses (367 of 793), only a small volume got discarded — 2.9% per dose, on average.
Multiplying unused milligrams by the cost per milligram, Dr. Sarfaty and colleagues estimated that, for each patient, $3,127 of the drug got discarded. When calculated over the year, the cost came to just over $200,000 at MSK, and nearly $15 million when projected across the approved patient population in the United States.
“Ultimately, we did not see a lot of waste with this specific drug,” Dr. Sarfaty said. “Under 2.9% is considered a reasonable amount, below the 3% threshold Peter Bach, MD, and colleagues recommend. But even with this small amount of waste, the cost per patient and to the system remains notable.”
The problem with recouping drug waste
Estimates from the Centers for Medicare & Medicaid Services (CMS), which tracks costs associated with discarded weight-based drugs covered under Medicare Part B, support the notion that small quantities of discarded drugs can still translate to big bucks.
Since 2017, CMS has required healthcare providers to report the volume of drugs discarded from a single-dose vial using a code, known as the JW modifier. The JW modifier means that providers can be reimbursed for the entire vial amount, not just the quantity the patient used.
In 2019, claims data from Medicare Part B showed that 1.85% of discarded rituximab came to $33.3 million. For infliximab, the 1.55% of discarded liquid translated to $15 million, and just 0.36% of discarded pembrolizumab reached $10 million.
However, experts question whether the JW modifier accurately reflects the quantity of drugs discarded.
According to the 2021 report from the National Academies of Sciences, Engineering, and Medicine (NASEM), most physicians don’t use the JW modifier. Among Medicare claims, 16.2% included the JW modifier in 2017 and 16.9% did in 2018.
The rate was significantly lower for private insurance. Of more than 4 million private insurance claims on 77 drugs made in 2017 and 2018, only 3.6% included the JW modifier; 15 of these drugs had no JW claims.
“Although we found that most physicians don’t use the JW modifier, even those who do, don’t use it consistently, even for the same patient,” said Dr. Yabroff, a co-author on the report.
Going a step further, Dr. Yabroff and colleagues argue that even if everyone used the JW modifier as intended, manufacturers would probably increase the price of drugs to compensate for any loss, potentially eliminating savings for payers.
That’s because, in the United States, manufacturers typically base drug prices on a patient and payers’ “willingness to pay for better health,” not on the volume of liquid used. Take a patient who pays $2,000 to receive the dose they need. If that dose is 600 mg but requires using two vials of 400 mg, then “to the patient, the 600-mg dose is worth $2,000, and the remainder has no value whatsoever,” the NASEM authors argue.
The authors parallel this scenario to purchasing a designer coat or dress. If that item requires alterations that remove a section of material, “the customer does not typically get a rebate because all the fabric was not needed,” the NASEM team writes.
But there’s a flaw in this rationale, argues Daniel Goldstein, MD, a medical oncologist at the Rabin Medical Center, Petah Tikva, Israel. A person’s willingness to pay for better health assumes that the price of a drug is based on proper market forces, where a drug’s cost and its effectiveness are in harmony.
“The problem is we’re operating in a broken market where the prices of oncology drugs have no real bearing on their efficacy,” said Dr. Goldstein.
And, as Dr. Bach noted in a 2021 Health Affairs piece, willingness to pay also requires that consumers know what they’re paying and allows them to walk away from an excessively high price.
But neither is a reality.
For one, Dr. Bach explains, companies may lowball the monthly price of a drug. In 2020, GlaxoSmithKline (GSK) announced that its new drug Blenrep would carry a list price of $8,277 per vial, or about $23,900 per month for an average 79 kg (175 lb) patient. That price accounts for two vials of the drug. But, according to Dr. Bach, “what GSK left out is that 44% of U.S. adults weigh more than 80 kg, and above that weight, three vials are needed per dose.” That would raise the average monthly cost to $30,479.
Perhaps more importantly, consumers can’t easily walk away.
“Medicare can’t negotiate prices and is forced to pay what a drug company says,” Dr. Goldstein said. “This is very different to when I buy a coat. If the price is too high, I can walk away.”
Fixed dosing: A solution or a new problem?
Efforts to reduce the financial impact of discarded cancer drugs can blow back on physicians, patients, and payers in other unanticipated ways. Take fixed dosing. Although chemotherapy dosing remains weight-based, many targeted therapies — such as nivolumab and pembrolizumab — recently transitioned to a fixed dosing regimen.
Administering a fixed, instead of weight-based, dose eliminates waste but can create new problems.
“Patients with cancer not only tend to get too high a dose of the drug, but costs go up significantly,” said Dr. Goldstein. In a 2017 analysis, Dr. Goldstein and colleagues compared dosing strategies in patients with metastatic non–small cell lung cancer who received pembrolizumab. The team found that the total annual cost of weight-based dosing was $2.6 billion, whereas the cost of the fixed dosing strategy was $3.44 billion — 24% more. In other words, personalized weight-based dosing would save more than $825 million dollars in the United States each year.
A 2020 analysis based in France found a similar cost increase of 26% for fixed dosing of pembrolizumab as well as nivolumab.
“I’ve argued we should go back to weight-based dosing,” Dr. Goldstein said. “Why should we give a higher dose with the same efficacy when that dose will cost significantly more and has the potential to increase adverse events?”
Does dose rounding work?
Rose DiMarco, PharmD, BCPS, BCOP, keeps a tight watch on patients being treated at the Sidney Kimmel Cancer Center at Jefferson Health in Philadelphia.
Dr. DiMarco educates patients about their treatment plan, reviews their lab results, and monitors them for side effects and drug interactions.
She also thinks a lot about costs.
“We spend about $100,000 a day on oncology drugs, and we want to make sure we’re not being wasteful,” Dr. DiMarco said in an interview.
One major initiative to curb waste and reduce costs at Jefferson has centered on dose rounding, which calculates whether a specific dose can be altered slightly to conserve vials and prevent waste. According to the Hematology/Oncology Pharmacy Association, a patient can receive up to 10% more or less of a weight-based dose without impacting treatment efficacy.
If, for instance, a patient requires 380 mg, but two vials come to 400 mg, rounding up that dose by approximately 5% means eliminating 20 mg that would go unused. But if that patient requires 420 mg, rounding down about 5% means substantial savings from not opening a new vial.
At Jefferson, Dr. DiMarco and her pharmacy colleagues map out dose ranges for all patients. Anyone who falls inside the 10% may be eligible for dose rounding. Anyone who doesn’t will receive the usual dose.
Although it is a challenge to implement, dose rounding has become standard of care at many cancer centers across the United States and is linked to substantial savings.
A 2018 analysis projected annual savings of $865,000 associated with rounding down eight monoclonal antibodies for patients with metastatic disease at a community cancer center. A more recent analysis from the Mayo Clinic found that dose rounding saved a total of 9,814 drug vials — 4485 of which were cancer drugs and 5329 of which were biologics — and resulted in $7.3 million in savings over 6 months in 2019 — $1.56 million from oncology agents and $5.7 from biologics.
And in a small 2019 analysis, researchers at Jefferson showed dose rounding of one monoclonal antibody saved approximately $30,000 in just 3 months, Dr. DiMarco noted.
“Not only does this process reduce costs and waste, but it also standardizes the preparation of hazardous medications, which can help prevent medication errors,” Dr. DiMarco said.
Nibbling around the edges
Despite estimates that scale into the billions of dollars, “drug wastage is just a small part of overall cancer costs,” Dr. Sarfaty said.
Fumiko Chino, MD, a radiation oncologist at MSK, agrees. “When we talk about affordability and cost, we can nibble around the edges of what’s really important,” Dr. Chino said. “Discarded drugs may cost a lot when you consider them in aggregate, but they are not as important as negotiated drug prices, which could substantially reduce overall costs.”
And until drug prices are addressed on a broader policy level, the cost of cancer care likely won’t improve in a meaningful way.
“But for the patient sitting in front of me, my focus will always be to provide the best care possible,” Dr. Binder said.
A version of this article first appeared on Medscape.com.
Three billion dollars: It’s enough to finance the annual out-of-pocket costs for 1 in 7 patients with cancer. It would cover almost half of the National Cancer Institute’s annual budget. And it could fund President Biden’s entire Cancer Moonshot program, with more than a billion to spare.
It’s also how much the United States spends on unused cancer drugs each year, some experts estimate.
Drug companies typically sell infused drugs in one or two single-dose vial sizes, but patients don’t come in such neat packages. A patient may need 300 mg of a drug that is only sold as 200 mg vials, which means half of a vial will go to waste.
Although most oncology drugs don’t incur substantial waste, even small volumes can translate to millions of dollars a year.
But can this money be saved or reallocated, if only we delivered drugs more efficiently?
Some experts don’t believe that’s possible.
“Attempts to recoup money for discarded drugs wouldn’t happen in a vacuum,” said Robin Yabroff, PhD, MBA, an epidemiologist and scientific vice president of Health Services Research at the American Cancer Society, who was part of a committee commissioned to evaluate the costs associated with discarded drugs.
The potential catch of any widespread effort to seek repayment or reduce the amount of discarded drugs, Dr. Yabroff and colleagues note, is that manufacturers would “simply increase the price of the vial.”
In other words, attempting to fix one problem may lead to another — essentially a whack-a-mole of cancer costs, which are projected to balloon to $246 billion by 2030.
What this means is without sweeping policies to rein in cancer care costs, oncologists can only do so much. And every little bit counts.
“We are left chipping away at this monster of cancer care costs,” said Adam Binder, MD, a medical oncologist at the Sidney Kimmel Cancer Center at Jefferson Health in Philadelphia.
Millions spent on “reasonable amount” of waste
Michal Sarfaty, MD, was excited when enfortumab vedotin came on the market to treat advanced urothelial cancer in late 2019.
The cost of the drug, however, tempered her enthusiasm.
Enfortumab vedotin is a “great drug,” said Dr. Sarfaty, an oncologist at the Sheba Medical Center, Ramat Gan, Israel. But it can cost upwards of $500,000 a year for an average-weight man.
Given the expense, Dr. Sarfaty wanted to understand how much of the drug gets thrown away. During a fellowship at Memorial Sloan Kettering (MSK) Cancer Center in New York, Dr. Sarfaty explored the amount of unused enfortumab vedotin among the 64 patients who received the drug in 2020. She, along with a team at MSK, calculated the price tag of that waste and extrapolated those estimates for patients across the country.
Although waste occurred in almost half of administered doses (367 of 793), only a small volume got discarded — 2.9% per dose, on average.
Multiplying unused milligrams by the cost per milligram, Dr. Sarfaty and colleagues estimated that, for each patient, $3,127 of the drug got discarded. When calculated over the year, the cost came to just over $200,000 at MSK, and nearly $15 million when projected across the approved patient population in the United States.
“Ultimately, we did not see a lot of waste with this specific drug,” Dr. Sarfaty said. “Under 2.9% is considered a reasonable amount, below the 3% threshold Peter Bach, MD, and colleagues recommend. But even with this small amount of waste, the cost per patient and to the system remains notable.”
The problem with recouping drug waste
Estimates from the Centers for Medicare & Medicaid Services (CMS), which tracks costs associated with discarded weight-based drugs covered under Medicare Part B, support the notion that small quantities of discarded drugs can still translate to big bucks.
Since 2017, CMS has required healthcare providers to report the volume of drugs discarded from a single-dose vial using a code, known as the JW modifier. The JW modifier means that providers can be reimbursed for the entire vial amount, not just the quantity the patient used.
In 2019, claims data from Medicare Part B showed that 1.85% of discarded rituximab came to $33.3 million. For infliximab, the 1.55% of discarded liquid translated to $15 million, and just 0.36% of discarded pembrolizumab reached $10 million.
However, experts question whether the JW modifier accurately reflects the quantity of drugs discarded.
According to the 2021 report from the National Academies of Sciences, Engineering, and Medicine (NASEM), most physicians don’t use the JW modifier. Among Medicare claims, 16.2% included the JW modifier in 2017 and 16.9% did in 2018.
The rate was significantly lower for private insurance. Of more than 4 million private insurance claims on 77 drugs made in 2017 and 2018, only 3.6% included the JW modifier; 15 of these drugs had no JW claims.
“Although we found that most physicians don’t use the JW modifier, even those who do, don’t use it consistently, even for the same patient,” said Dr. Yabroff, a co-author on the report.
Going a step further, Dr. Yabroff and colleagues argue that even if everyone used the JW modifier as intended, manufacturers would probably increase the price of drugs to compensate for any loss, potentially eliminating savings for payers.
That’s because, in the United States, manufacturers typically base drug prices on a patient and payers’ “willingness to pay for better health,” not on the volume of liquid used. Take a patient who pays $2,000 to receive the dose they need. If that dose is 600 mg but requires using two vials of 400 mg, then “to the patient, the 600-mg dose is worth $2,000, and the remainder has no value whatsoever,” the NASEM authors argue.
The authors parallel this scenario to purchasing a designer coat or dress. If that item requires alterations that remove a section of material, “the customer does not typically get a rebate because all the fabric was not needed,” the NASEM team writes.
But there’s a flaw in this rationale, argues Daniel Goldstein, MD, a medical oncologist at the Rabin Medical Center, Petah Tikva, Israel. A person’s willingness to pay for better health assumes that the price of a drug is based on proper market forces, where a drug’s cost and its effectiveness are in harmony.
“The problem is we’re operating in a broken market where the prices of oncology drugs have no real bearing on their efficacy,” said Dr. Goldstein.
And, as Dr. Bach noted in a 2021 Health Affairs piece, willingness to pay also requires that consumers know what they’re paying and allows them to walk away from an excessively high price.
But neither is a reality.
For one, Dr. Bach explains, companies may lowball the monthly price of a drug. In 2020, GlaxoSmithKline (GSK) announced that its new drug Blenrep would carry a list price of $8,277 per vial, or about $23,900 per month for an average 79 kg (175 lb) patient. That price accounts for two vials of the drug. But, according to Dr. Bach, “what GSK left out is that 44% of U.S. adults weigh more than 80 kg, and above that weight, three vials are needed per dose.” That would raise the average monthly cost to $30,479.
Perhaps more importantly, consumers can’t easily walk away.
“Medicare can’t negotiate prices and is forced to pay what a drug company says,” Dr. Goldstein said. “This is very different to when I buy a coat. If the price is too high, I can walk away.”
Fixed dosing: A solution or a new problem?
Efforts to reduce the financial impact of discarded cancer drugs can blow back on physicians, patients, and payers in other unanticipated ways. Take fixed dosing. Although chemotherapy dosing remains weight-based, many targeted therapies — such as nivolumab and pembrolizumab — recently transitioned to a fixed dosing regimen.
Administering a fixed, instead of weight-based, dose eliminates waste but can create new problems.
“Patients with cancer not only tend to get too high a dose of the drug, but costs go up significantly,” said Dr. Goldstein. In a 2017 analysis, Dr. Goldstein and colleagues compared dosing strategies in patients with metastatic non–small cell lung cancer who received pembrolizumab. The team found that the total annual cost of weight-based dosing was $2.6 billion, whereas the cost of the fixed dosing strategy was $3.44 billion — 24% more. In other words, personalized weight-based dosing would save more than $825 million dollars in the United States each year.
A 2020 analysis based in France found a similar cost increase of 26% for fixed dosing of pembrolizumab as well as nivolumab.
“I’ve argued we should go back to weight-based dosing,” Dr. Goldstein said. “Why should we give a higher dose with the same efficacy when that dose will cost significantly more and has the potential to increase adverse events?”
Does dose rounding work?
Rose DiMarco, PharmD, BCPS, BCOP, keeps a tight watch on patients being treated at the Sidney Kimmel Cancer Center at Jefferson Health in Philadelphia.
Dr. DiMarco educates patients about their treatment plan, reviews their lab results, and monitors them for side effects and drug interactions.
She also thinks a lot about costs.
“We spend about $100,000 a day on oncology drugs, and we want to make sure we’re not being wasteful,” Dr. DiMarco said in an interview.
One major initiative to curb waste and reduce costs at Jefferson has centered on dose rounding, which calculates whether a specific dose can be altered slightly to conserve vials and prevent waste. According to the Hematology/Oncology Pharmacy Association, a patient can receive up to 10% more or less of a weight-based dose without impacting treatment efficacy.
If, for instance, a patient requires 380 mg, but two vials come to 400 mg, rounding up that dose by approximately 5% means eliminating 20 mg that would go unused. But if that patient requires 420 mg, rounding down about 5% means substantial savings from not opening a new vial.
At Jefferson, Dr. DiMarco and her pharmacy colleagues map out dose ranges for all patients. Anyone who falls inside the 10% may be eligible for dose rounding. Anyone who doesn’t will receive the usual dose.
Although it is a challenge to implement, dose rounding has become standard of care at many cancer centers across the United States and is linked to substantial savings.
A 2018 analysis projected annual savings of $865,000 associated with rounding down eight monoclonal antibodies for patients with metastatic disease at a community cancer center. A more recent analysis from the Mayo Clinic found that dose rounding saved a total of 9,814 drug vials — 4485 of which were cancer drugs and 5329 of which were biologics — and resulted in $7.3 million in savings over 6 months in 2019 — $1.56 million from oncology agents and $5.7 from biologics.
And in a small 2019 analysis, researchers at Jefferson showed dose rounding of one monoclonal antibody saved approximately $30,000 in just 3 months, Dr. DiMarco noted.
“Not only does this process reduce costs and waste, but it also standardizes the preparation of hazardous medications, which can help prevent medication errors,” Dr. DiMarco said.
Nibbling around the edges
Despite estimates that scale into the billions of dollars, “drug wastage is just a small part of overall cancer costs,” Dr. Sarfaty said.
Fumiko Chino, MD, a radiation oncologist at MSK, agrees. “When we talk about affordability and cost, we can nibble around the edges of what’s really important,” Dr. Chino said. “Discarded drugs may cost a lot when you consider them in aggregate, but they are not as important as negotiated drug prices, which could substantially reduce overall costs.”
And until drug prices are addressed on a broader policy level, the cost of cancer care likely won’t improve in a meaningful way.
“But for the patient sitting in front of me, my focus will always be to provide the best care possible,” Dr. Binder said.
A version of this article first appeared on Medscape.com.
Racial disparities in endometrial cancer
Endometrial cancer (EC) is the most common gynecologic malignancy and is the fourth most common cancer seen in U.S. women. It is the only major cancer that has continued to see a rise in incidence and mortality for the past 2 decades, and it is anticipated that nearly 66,000 new cases of EC will be diagnosed this year with 12,550 deaths.1 Given that the well-established risk factors for developing EC including obesity, diabetes, and insulin resistance, the obesity epidemic is indisputably playing a significant role in the increasing incidence.
Historically, White women were thought to have the highest incidence of EC; however, this incidence rate did not account for hysterectomy prevalence, which can vary widely by numerous factors including age, race, ethnicity, and geographic region. When correcting EC incidence rates for prevalence of hysterectomy, Black women have had the highest incidence of EC since 2007, and rates continue to climb.2 In fact, the average annual percent change (APC) in EC incidence from 2000 to 2015 was stable for White women at 0.2% while Black women had a near order of magnitude greater APC at 2.1%.2
Differing incidence rates of EC can also be seen by histologic subtype. Endometrioid EC is the more common and less lethal histology of EC that often coincides with the type I classification of EC. These tumors are estrogen driven; therefore, they are associated with conditions resulting in excess estrogen (for example, anovulation, obesity, and hyperlipidemia). Nonendometrioid histologies, primarily composed of serous tumors, are more rare, are typically more aggressive, are not estrogen driven, and are commonly classified as type II tumors. Racial differences between type I and type II tumors are seen with White women more commonly being diagnosed with type I tumors while Black women more typically have type II tumors. White women have the greatest incidence rate of endometrioid EC with an APC that remained relatively unchanged from 2000 to 2015. Black women’s APC in incidence rate of endometrioid EC has increased during this same period at 1.3%. For nonendometrioid tumors, an increasing incidence is seen in all races and ethnicities; however, Black women have a much higher incidence of these tumors, with a rate that continues to increase at an APC of 3.2%.2
EC incidence is increasing with a particularly concerning rise in those who report Black race, but are these same disparities being seen in EC mortality? Unfortunately, drastic disparities are seen in survival data for Black women afflicted with EC. Black patients are more likely to be diagnosed with advanced or metastatic EC and less likely to be diagnosed with localized tumors. While being diagnosed with a more advanced stage of disease does affect survival in EC, Black patients have worse survival regardless of stage of disease at the time of diagnosis.1 As discussed earlier, the more aggressive type II tumors are composed of nonendometrioid histologies and are more common in Black women. This could lead to the false assumption that these higher-risk tumors are why Black women are disproportionately dying from EC; however, when examining survival by histologic subtype, Black women are more frequently dying from the lower-risk endometrioid EC regardless of stage of disease. The same disparate survival outcomes are also seen in nonendometrioid histologies.2 Thus, Black patients have the lowest survival rates irrespective of stage at diagnosis or histologic subtype.
The disparities seen in EC mortality are not new. They can be seen in data for over 30 years and are only widening. While there has been an increase in mortality rates from EC across all races and ethnicities from 2015 to 2019 compared with 1990 to 1994, the mortality rate ratio for Black women compared with White women has increased from 1.83 in 1990-1994 to 1.98 in 2015-2019.3 In the early 1990s, the risk of death from ovarian cancer was twice that of EC. The mortality of EC is now similar to that of ovarian cancer. This threshold in mortality ratio of EC to ovarian cancer has already been seen in Black women, who have experienced greater mortality in EC compared with ovarian cancer since 2005. In fact, the EC mortality of Black women in 2019 was similar to the mortality of White women with ovarian cancer nearly 30 years ago.3
Decades of data have demonstrated the glaring racial disparities seen in EC, and yet, no significant progress has been made in addressing this inequity. Oncology research is now beginning to move beyond describing these differences to a strategy of achieving equitable cancer care. While the study frameworks and novel investigations aimed at addressing the disparities in EC is outside the scope of this article, disparities in clinical trial enrollment continue to exist.
A recent example can be seen in the practice-changing KEYNOTE-775 trial, which led to the Food and Drug Administration approval of lenvatinib plus pembrolizumab in EC treatment.4 A total of 827 patients with EC that progressed or recurred following treatment with platinum-based chemotherapy were enrolled in this multinational, multicenter trial. Thirty-one (3.7%) of the patients enrolled were Black. Of those who were enrolled in the United States, 14% were Black. The authors report that this proportion of Black patients in the United States is consistent with 2020 census data, which reported 13.4% of people identified as Black. However, using census data as a benchmark for equitable enrollment is inappropriate. Certain demographic groups are historically more difficult to count, and the COVID-19 pandemic exacerbated the challenge in obtaining an accurate count through job loss, government distrust, and access restrictions resulting in an estimated net undercount of 2.45% in those who report Black race.5 Composition of trial enrollment should mirror the population that will be affected by the study results. As advanced EC disproportionately affects Black patients, their enrollment must be higher in these pivotal trials. How else are we to know if these novel therapeutics will work in the population that is most afflicted by EC?
Future studies must account for socioeconomic factors while acknowledging the role of social determinants of health. It is imperative that we use the knowledge that race is a social construct created to control access to power and that there are biologic responses to environmental stresses, including that of racism, affecting health and disease. Changes at every level, from individual practitioners up to federal policies, will need to be enacted or else the unacceptable status quo will continue.
Dr. Burkett is a clinical fellow in the division of gynecologic oncology, department of obstetrics and gynecology, at the University of North Carolina at Chapel Hill.
References
1. Siegel RL et al. CA Cancer J Clin. 2022;72:7-33.
2. Clarke MA et al. J Clin Oncol. 2019;37:1895-908.
3. Giaquinto AN et al. Obstet Gynecol. 2022;139:440-2.
4. Makker V et al. N Engl J Med. 2022;386:437-48.
5. Elliott D et al. Simulating the 2020 Census: Miscounts and the fairness of outcomes. Urban Institute; 2021.
Endometrial cancer (EC) is the most common gynecologic malignancy and is the fourth most common cancer seen in U.S. women. It is the only major cancer that has continued to see a rise in incidence and mortality for the past 2 decades, and it is anticipated that nearly 66,000 new cases of EC will be diagnosed this year with 12,550 deaths.1 Given that the well-established risk factors for developing EC including obesity, diabetes, and insulin resistance, the obesity epidemic is indisputably playing a significant role in the increasing incidence.
Historically, White women were thought to have the highest incidence of EC; however, this incidence rate did not account for hysterectomy prevalence, which can vary widely by numerous factors including age, race, ethnicity, and geographic region. When correcting EC incidence rates for prevalence of hysterectomy, Black women have had the highest incidence of EC since 2007, and rates continue to climb.2 In fact, the average annual percent change (APC) in EC incidence from 2000 to 2015 was stable for White women at 0.2% while Black women had a near order of magnitude greater APC at 2.1%.2
Differing incidence rates of EC can also be seen by histologic subtype. Endometrioid EC is the more common and less lethal histology of EC that often coincides with the type I classification of EC. These tumors are estrogen driven; therefore, they are associated with conditions resulting in excess estrogen (for example, anovulation, obesity, and hyperlipidemia). Nonendometrioid histologies, primarily composed of serous tumors, are more rare, are typically more aggressive, are not estrogen driven, and are commonly classified as type II tumors. Racial differences between type I and type II tumors are seen with White women more commonly being diagnosed with type I tumors while Black women more typically have type II tumors. White women have the greatest incidence rate of endometrioid EC with an APC that remained relatively unchanged from 2000 to 2015. Black women’s APC in incidence rate of endometrioid EC has increased during this same period at 1.3%. For nonendometrioid tumors, an increasing incidence is seen in all races and ethnicities; however, Black women have a much higher incidence of these tumors, with a rate that continues to increase at an APC of 3.2%.2
EC incidence is increasing with a particularly concerning rise in those who report Black race, but are these same disparities being seen in EC mortality? Unfortunately, drastic disparities are seen in survival data for Black women afflicted with EC. Black patients are more likely to be diagnosed with advanced or metastatic EC and less likely to be diagnosed with localized tumors. While being diagnosed with a more advanced stage of disease does affect survival in EC, Black patients have worse survival regardless of stage of disease at the time of diagnosis.1 As discussed earlier, the more aggressive type II tumors are composed of nonendometrioid histologies and are more common in Black women. This could lead to the false assumption that these higher-risk tumors are why Black women are disproportionately dying from EC; however, when examining survival by histologic subtype, Black women are more frequently dying from the lower-risk endometrioid EC regardless of stage of disease. The same disparate survival outcomes are also seen in nonendometrioid histologies.2 Thus, Black patients have the lowest survival rates irrespective of stage at diagnosis or histologic subtype.
The disparities seen in EC mortality are not new. They can be seen in data for over 30 years and are only widening. While there has been an increase in mortality rates from EC across all races and ethnicities from 2015 to 2019 compared with 1990 to 1994, the mortality rate ratio for Black women compared with White women has increased from 1.83 in 1990-1994 to 1.98 in 2015-2019.3 In the early 1990s, the risk of death from ovarian cancer was twice that of EC. The mortality of EC is now similar to that of ovarian cancer. This threshold in mortality ratio of EC to ovarian cancer has already been seen in Black women, who have experienced greater mortality in EC compared with ovarian cancer since 2005. In fact, the EC mortality of Black women in 2019 was similar to the mortality of White women with ovarian cancer nearly 30 years ago.3
Decades of data have demonstrated the glaring racial disparities seen in EC, and yet, no significant progress has been made in addressing this inequity. Oncology research is now beginning to move beyond describing these differences to a strategy of achieving equitable cancer care. While the study frameworks and novel investigations aimed at addressing the disparities in EC is outside the scope of this article, disparities in clinical trial enrollment continue to exist.
A recent example can be seen in the practice-changing KEYNOTE-775 trial, which led to the Food and Drug Administration approval of lenvatinib plus pembrolizumab in EC treatment.4 A total of 827 patients with EC that progressed or recurred following treatment with platinum-based chemotherapy were enrolled in this multinational, multicenter trial. Thirty-one (3.7%) of the patients enrolled were Black. Of those who were enrolled in the United States, 14% were Black. The authors report that this proportion of Black patients in the United States is consistent with 2020 census data, which reported 13.4% of people identified as Black. However, using census data as a benchmark for equitable enrollment is inappropriate. Certain demographic groups are historically more difficult to count, and the COVID-19 pandemic exacerbated the challenge in obtaining an accurate count through job loss, government distrust, and access restrictions resulting in an estimated net undercount of 2.45% in those who report Black race.5 Composition of trial enrollment should mirror the population that will be affected by the study results. As advanced EC disproportionately affects Black patients, their enrollment must be higher in these pivotal trials. How else are we to know if these novel therapeutics will work in the population that is most afflicted by EC?
Future studies must account for socioeconomic factors while acknowledging the role of social determinants of health. It is imperative that we use the knowledge that race is a social construct created to control access to power and that there are biologic responses to environmental stresses, including that of racism, affecting health and disease. Changes at every level, from individual practitioners up to federal policies, will need to be enacted or else the unacceptable status quo will continue.
Dr. Burkett is a clinical fellow in the division of gynecologic oncology, department of obstetrics and gynecology, at the University of North Carolina at Chapel Hill.
References
1. Siegel RL et al. CA Cancer J Clin. 2022;72:7-33.
2. Clarke MA et al. J Clin Oncol. 2019;37:1895-908.
3. Giaquinto AN et al. Obstet Gynecol. 2022;139:440-2.
4. Makker V et al. N Engl J Med. 2022;386:437-48.
5. Elliott D et al. Simulating the 2020 Census: Miscounts and the fairness of outcomes. Urban Institute; 2021.
Endometrial cancer (EC) is the most common gynecologic malignancy and is the fourth most common cancer seen in U.S. women. It is the only major cancer that has continued to see a rise in incidence and mortality for the past 2 decades, and it is anticipated that nearly 66,000 new cases of EC will be diagnosed this year with 12,550 deaths.1 Given that the well-established risk factors for developing EC including obesity, diabetes, and insulin resistance, the obesity epidemic is indisputably playing a significant role in the increasing incidence.
Historically, White women were thought to have the highest incidence of EC; however, this incidence rate did not account for hysterectomy prevalence, which can vary widely by numerous factors including age, race, ethnicity, and geographic region. When correcting EC incidence rates for prevalence of hysterectomy, Black women have had the highest incidence of EC since 2007, and rates continue to climb.2 In fact, the average annual percent change (APC) in EC incidence from 2000 to 2015 was stable for White women at 0.2% while Black women had a near order of magnitude greater APC at 2.1%.2
Differing incidence rates of EC can also be seen by histologic subtype. Endometrioid EC is the more common and less lethal histology of EC that often coincides with the type I classification of EC. These tumors are estrogen driven; therefore, they are associated with conditions resulting in excess estrogen (for example, anovulation, obesity, and hyperlipidemia). Nonendometrioid histologies, primarily composed of serous tumors, are more rare, are typically more aggressive, are not estrogen driven, and are commonly classified as type II tumors. Racial differences between type I and type II tumors are seen with White women more commonly being diagnosed with type I tumors while Black women more typically have type II tumors. White women have the greatest incidence rate of endometrioid EC with an APC that remained relatively unchanged from 2000 to 2015. Black women’s APC in incidence rate of endometrioid EC has increased during this same period at 1.3%. For nonendometrioid tumors, an increasing incidence is seen in all races and ethnicities; however, Black women have a much higher incidence of these tumors, with a rate that continues to increase at an APC of 3.2%.2
EC incidence is increasing with a particularly concerning rise in those who report Black race, but are these same disparities being seen in EC mortality? Unfortunately, drastic disparities are seen in survival data for Black women afflicted with EC. Black patients are more likely to be diagnosed with advanced or metastatic EC and less likely to be diagnosed with localized tumors. While being diagnosed with a more advanced stage of disease does affect survival in EC, Black patients have worse survival regardless of stage of disease at the time of diagnosis.1 As discussed earlier, the more aggressive type II tumors are composed of nonendometrioid histologies and are more common in Black women. This could lead to the false assumption that these higher-risk tumors are why Black women are disproportionately dying from EC; however, when examining survival by histologic subtype, Black women are more frequently dying from the lower-risk endometrioid EC regardless of stage of disease. The same disparate survival outcomes are also seen in nonendometrioid histologies.2 Thus, Black patients have the lowest survival rates irrespective of stage at diagnosis or histologic subtype.
The disparities seen in EC mortality are not new. They can be seen in data for over 30 years and are only widening. While there has been an increase in mortality rates from EC across all races and ethnicities from 2015 to 2019 compared with 1990 to 1994, the mortality rate ratio for Black women compared with White women has increased from 1.83 in 1990-1994 to 1.98 in 2015-2019.3 In the early 1990s, the risk of death from ovarian cancer was twice that of EC. The mortality of EC is now similar to that of ovarian cancer. This threshold in mortality ratio of EC to ovarian cancer has already been seen in Black women, who have experienced greater mortality in EC compared with ovarian cancer since 2005. In fact, the EC mortality of Black women in 2019 was similar to the mortality of White women with ovarian cancer nearly 30 years ago.3
Decades of data have demonstrated the glaring racial disparities seen in EC, and yet, no significant progress has been made in addressing this inequity. Oncology research is now beginning to move beyond describing these differences to a strategy of achieving equitable cancer care. While the study frameworks and novel investigations aimed at addressing the disparities in EC is outside the scope of this article, disparities in clinical trial enrollment continue to exist.
A recent example can be seen in the practice-changing KEYNOTE-775 trial, which led to the Food and Drug Administration approval of lenvatinib plus pembrolizumab in EC treatment.4 A total of 827 patients with EC that progressed or recurred following treatment with platinum-based chemotherapy were enrolled in this multinational, multicenter trial. Thirty-one (3.7%) of the patients enrolled were Black. Of those who were enrolled in the United States, 14% were Black. The authors report that this proportion of Black patients in the United States is consistent with 2020 census data, which reported 13.4% of people identified as Black. However, using census data as a benchmark for equitable enrollment is inappropriate. Certain demographic groups are historically more difficult to count, and the COVID-19 pandemic exacerbated the challenge in obtaining an accurate count through job loss, government distrust, and access restrictions resulting in an estimated net undercount of 2.45% in those who report Black race.5 Composition of trial enrollment should mirror the population that will be affected by the study results. As advanced EC disproportionately affects Black patients, their enrollment must be higher in these pivotal trials. How else are we to know if these novel therapeutics will work in the population that is most afflicted by EC?
Future studies must account for socioeconomic factors while acknowledging the role of social determinants of health. It is imperative that we use the knowledge that race is a social construct created to control access to power and that there are biologic responses to environmental stresses, including that of racism, affecting health and disease. Changes at every level, from individual practitioners up to federal policies, will need to be enacted or else the unacceptable status quo will continue.
Dr. Burkett is a clinical fellow in the division of gynecologic oncology, department of obstetrics and gynecology, at the University of North Carolina at Chapel Hill.
References
1. Siegel RL et al. CA Cancer J Clin. 2022;72:7-33.
2. Clarke MA et al. J Clin Oncol. 2019;37:1895-908.
3. Giaquinto AN et al. Obstet Gynecol. 2022;139:440-2.
4. Makker V et al. N Engl J Med. 2022;386:437-48.
5. Elliott D et al. Simulating the 2020 Census: Miscounts and the fairness of outcomes. Urban Institute; 2021.
Migraine-related stigma is common and underappreciated
, new research shows. Results from the OVERCOME population-based survey study, which included more than 59,000 respondents, showed about 32% reported experiencing migraine-related stigma “often” or “very often.”
Even those experiencing only a few headaches per month said they experienced negative attitudes from others about migraine.
“We have been utterly blind to the burden people with migraine experience in terms of how their disease is appreciated by others. It’s time to get busy and address this very stubborn social phenomenon,” said the study’s coinvestigator, Robert E. Shapiro, MD, PhD, professor emeritus, department of neurological sciences, Larner College of Medicine, University of Vermont, Burlington.
The findings were presented at the annual meeting of the American Headache Society.
Population-based research
Stigma is defined as the discounting or discrediting of an individual with a trait that deviates from social norms. To date, there have been no significant population-based studies of how these attitudes affect individuals with migraine.
OVERCOME is a cross-sectional, longitudinal, prospective, web-based survey conducted in a representative sample of the United States population. For this new analysis, researchers pooled data from surveys conducted in 2018, 2019, and 2020.
The analysis included 59,004 respondents (mean age, 41.3 years; 75% women; 70% White) who reported one or more headache or migraine attacks during the previous 12 months and who met criteria for migraine from the International Classification of Headache Disorders. Among the patients, 35% had a college degree, and 89% suffered episodic-type headaches.
Researchers used the following four patient-reported outcome measures:
- Migraine Disability Assessment, which quantifies number of days missed at work, home, or social events over the previous 3 months
- Migraine Interictal Burden Scale–4, which measures burden of migraine between attacks over the previous 4 weeks
- Migraine-Specific Quality of Life Role-Function Restrictive, which assesses the functional effect of migraine on social and work-related activities over the previous 4 weeks
- Migraine-Related Stigma
For the latter, the investigators used two measures – the degree to which others think migraine is used to acquire secondary gain, such as avoiding commitments, and the degree to which others minimize the burden of migraine.
One of the most stigmatizing disorders
Results showed that 31.7% of participants reported experiencing stigmatization from one or both migraine-related stigma categories often or very often – a result Dr. Shapiro characterized as “pretty shocking.”
Participants with chronic headaches, defined as having 15 or more headache days per month, made up 11% of the sample. About 47% of these respondents felt stigma often or very often.
However, even 25% of participants with three or fewer headache days per month reported stigma.
“This is a fundamental tip that we are not really understanding the concerns that drive burden for people living with this disabling disease,” Dr. Shapiro said.
Some previous studies that compared levels of stigmatizing attitudes regarding various diseases showed that migraine is more stigmatized than even epilepsy, a condition “equated with demonic possession in biblical times,” he noted.
One study used machine learning to measure the number of pejorative or negative terms associated with various diseases. “Shockingly, migraine was one of the most stigmatized diseases,” said Dr. Shapiro. “It was as stigmatized as gonorrhea by certain measures.”
Results from the new study showed that, irrespective of the number of headache days experienced per month, there was a threefold increase in interictal burden among those reporting stigma often or very often, compared with those who didn’t report stigma.
Large impact on QoL
“Stigma is a social concept, so maybe it’s not surprising that it would be present whether or not someone is experiencing other symptoms of migraine,” said Dr. Shapiro.
Across all monthly headache days, experiencing more migraine-related stigma was associated with increased disability and decreased quality of life.
Dr. Shapiro noted that when it comes to migraine-specific quality of life, stigma appears to have more of an effect than number of headache days. “That means there are big gaps in our appreciation for what drives the burdens in the patient experience of living with this disabling disease,” he said.
He added that headache’s place within the migraine sphere needs to be reconsidered. “Its singular emphasis has limited our full appreciation of this disease and how we should be paying attention to the things that are important to patients,” he said.
Dr. Shapiro predicts that future clinical trials will include migraine-related stigma as a measure to guide trial enrollment.
In addition, researchers are now digging deeper to try to understand what factors are most likely to drive stigma. “We need to understand why those attitudes are held and who is more likely to hold those attitudes, and that may allow us to develop mitigating strategies to reduce those attitudes,” said Dr. Shapiro.
‘Worrisome’ findings
Commenting on the findings, Deborah Friedman, MD, professor, departments of neurology and of ophthalmology, UT Southwestern Medical Center, Dallas, said the data on stigma were “worrisome.”
Missing social occasions and lost productivity may make migraine more visible to others so perhaps may “provoke stigma or stigmatizing comments or stigmatizing attitudes,” said Dr. Friedman, who was not involved with the research.
She noted that patients with migraine might also have trouble functioning in the workplace. “They may not be able to tolerate the computer screen or smells of perfume at the office and not get accommodation for that,” she said.
In addition to external stigma, which was investigated in the study, individuals with migraine may also experience internal stigma – blaming themselves for their disease, which may make it less likely they will seek care, said Dr. Friedman.
“That’s a huge problem,” she added.
The OVERCOME study is funded by Lilly. Dr. Shapiro has been compensated by Lilly as a research consultant and as a member of the data monitoring committee for clinical trials for galcanezumab, a Lilly pharmaceutical. Dr. Friedman has reported no relevant financial relationships.
A version of this article first appeared on Medscape.com.
, new research shows. Results from the OVERCOME population-based survey study, which included more than 59,000 respondents, showed about 32% reported experiencing migraine-related stigma “often” or “very often.”
Even those experiencing only a few headaches per month said they experienced negative attitudes from others about migraine.
“We have been utterly blind to the burden people with migraine experience in terms of how their disease is appreciated by others. It’s time to get busy and address this very stubborn social phenomenon,” said the study’s coinvestigator, Robert E. Shapiro, MD, PhD, professor emeritus, department of neurological sciences, Larner College of Medicine, University of Vermont, Burlington.
The findings were presented at the annual meeting of the American Headache Society.
Population-based research
Stigma is defined as the discounting or discrediting of an individual with a trait that deviates from social norms. To date, there have been no significant population-based studies of how these attitudes affect individuals with migraine.
OVERCOME is a cross-sectional, longitudinal, prospective, web-based survey conducted in a representative sample of the United States population. For this new analysis, researchers pooled data from surveys conducted in 2018, 2019, and 2020.
The analysis included 59,004 respondents (mean age, 41.3 years; 75% women; 70% White) who reported one or more headache or migraine attacks during the previous 12 months and who met criteria for migraine from the International Classification of Headache Disorders. Among the patients, 35% had a college degree, and 89% suffered episodic-type headaches.
Researchers used the following four patient-reported outcome measures:
- Migraine Disability Assessment, which quantifies number of days missed at work, home, or social events over the previous 3 months
- Migraine Interictal Burden Scale–4, which measures burden of migraine between attacks over the previous 4 weeks
- Migraine-Specific Quality of Life Role-Function Restrictive, which assesses the functional effect of migraine on social and work-related activities over the previous 4 weeks
- Migraine-Related Stigma
For the latter, the investigators used two measures – the degree to which others think migraine is used to acquire secondary gain, such as avoiding commitments, and the degree to which others minimize the burden of migraine.
One of the most stigmatizing disorders
Results showed that 31.7% of participants reported experiencing stigmatization from one or both migraine-related stigma categories often or very often – a result Dr. Shapiro characterized as “pretty shocking.”
Participants with chronic headaches, defined as having 15 or more headache days per month, made up 11% of the sample. About 47% of these respondents felt stigma often or very often.
However, even 25% of participants with three or fewer headache days per month reported stigma.
“This is a fundamental tip that we are not really understanding the concerns that drive burden for people living with this disabling disease,” Dr. Shapiro said.
Some previous studies that compared levels of stigmatizing attitudes regarding various diseases showed that migraine is more stigmatized than even epilepsy, a condition “equated with demonic possession in biblical times,” he noted.
One study used machine learning to measure the number of pejorative or negative terms associated with various diseases. “Shockingly, migraine was one of the most stigmatized diseases,” said Dr. Shapiro. “It was as stigmatized as gonorrhea by certain measures.”
Results from the new study showed that, irrespective of the number of headache days experienced per month, there was a threefold increase in interictal burden among those reporting stigma often or very often, compared with those who didn’t report stigma.
Large impact on QoL
“Stigma is a social concept, so maybe it’s not surprising that it would be present whether or not someone is experiencing other symptoms of migraine,” said Dr. Shapiro.
Across all monthly headache days, experiencing more migraine-related stigma was associated with increased disability and decreased quality of life.
Dr. Shapiro noted that when it comes to migraine-specific quality of life, stigma appears to have more of an effect than number of headache days. “That means there are big gaps in our appreciation for what drives the burdens in the patient experience of living with this disabling disease,” he said.
He added that headache’s place within the migraine sphere needs to be reconsidered. “Its singular emphasis has limited our full appreciation of this disease and how we should be paying attention to the things that are important to patients,” he said.
Dr. Shapiro predicts that future clinical trials will include migraine-related stigma as a measure to guide trial enrollment.
In addition, researchers are now digging deeper to try to understand what factors are most likely to drive stigma. “We need to understand why those attitudes are held and who is more likely to hold those attitudes, and that may allow us to develop mitigating strategies to reduce those attitudes,” said Dr. Shapiro.
‘Worrisome’ findings
Commenting on the findings, Deborah Friedman, MD, professor, departments of neurology and of ophthalmology, UT Southwestern Medical Center, Dallas, said the data on stigma were “worrisome.”
Missing social occasions and lost productivity may make migraine more visible to others so perhaps may “provoke stigma or stigmatizing comments or stigmatizing attitudes,” said Dr. Friedman, who was not involved with the research.
She noted that patients with migraine might also have trouble functioning in the workplace. “They may not be able to tolerate the computer screen or smells of perfume at the office and not get accommodation for that,” she said.
In addition to external stigma, which was investigated in the study, individuals with migraine may also experience internal stigma – blaming themselves for their disease, which may make it less likely they will seek care, said Dr. Friedman.
“That’s a huge problem,” she added.
The OVERCOME study is funded by Lilly. Dr. Shapiro has been compensated by Lilly as a research consultant and as a member of the data monitoring committee for clinical trials for galcanezumab, a Lilly pharmaceutical. Dr. Friedman has reported no relevant financial relationships.
A version of this article first appeared on Medscape.com.
, new research shows. Results from the OVERCOME population-based survey study, which included more than 59,000 respondents, showed about 32% reported experiencing migraine-related stigma “often” or “very often.”
Even those experiencing only a few headaches per month said they experienced negative attitudes from others about migraine.
“We have been utterly blind to the burden people with migraine experience in terms of how their disease is appreciated by others. It’s time to get busy and address this very stubborn social phenomenon,” said the study’s coinvestigator, Robert E. Shapiro, MD, PhD, professor emeritus, department of neurological sciences, Larner College of Medicine, University of Vermont, Burlington.
The findings were presented at the annual meeting of the American Headache Society.
Population-based research
Stigma is defined as the discounting or discrediting of an individual with a trait that deviates from social norms. To date, there have been no significant population-based studies of how these attitudes affect individuals with migraine.
OVERCOME is a cross-sectional, longitudinal, prospective, web-based survey conducted in a representative sample of the United States population. For this new analysis, researchers pooled data from surveys conducted in 2018, 2019, and 2020.
The analysis included 59,004 respondents (mean age, 41.3 years; 75% women; 70% White) who reported one or more headache or migraine attacks during the previous 12 months and who met criteria for migraine from the International Classification of Headache Disorders. Among the patients, 35% had a college degree, and 89% suffered episodic-type headaches.
Researchers used the following four patient-reported outcome measures:
- Migraine Disability Assessment, which quantifies number of days missed at work, home, or social events over the previous 3 months
- Migraine Interictal Burden Scale–4, which measures burden of migraine between attacks over the previous 4 weeks
- Migraine-Specific Quality of Life Role-Function Restrictive, which assesses the functional effect of migraine on social and work-related activities over the previous 4 weeks
- Migraine-Related Stigma
For the latter, the investigators used two measures – the degree to which others think migraine is used to acquire secondary gain, such as avoiding commitments, and the degree to which others minimize the burden of migraine.
One of the most stigmatizing disorders
Results showed that 31.7% of participants reported experiencing stigmatization from one or both migraine-related stigma categories often or very often – a result Dr. Shapiro characterized as “pretty shocking.”
Participants with chronic headaches, defined as having 15 or more headache days per month, made up 11% of the sample. About 47% of these respondents felt stigma often or very often.
However, even 25% of participants with three or fewer headache days per month reported stigma.
“This is a fundamental tip that we are not really understanding the concerns that drive burden for people living with this disabling disease,” Dr. Shapiro said.
Some previous studies that compared levels of stigmatizing attitudes regarding various diseases showed that migraine is more stigmatized than even epilepsy, a condition “equated with demonic possession in biblical times,” he noted.
One study used machine learning to measure the number of pejorative or negative terms associated with various diseases. “Shockingly, migraine was one of the most stigmatized diseases,” said Dr. Shapiro. “It was as stigmatized as gonorrhea by certain measures.”
Results from the new study showed that, irrespective of the number of headache days experienced per month, there was a threefold increase in interictal burden among those reporting stigma often or very often, compared with those who didn’t report stigma.
Large impact on QoL
“Stigma is a social concept, so maybe it’s not surprising that it would be present whether or not someone is experiencing other symptoms of migraine,” said Dr. Shapiro.
Across all monthly headache days, experiencing more migraine-related stigma was associated with increased disability and decreased quality of life.
Dr. Shapiro noted that when it comes to migraine-specific quality of life, stigma appears to have more of an effect than number of headache days. “That means there are big gaps in our appreciation for what drives the burdens in the patient experience of living with this disabling disease,” he said.
He added that headache’s place within the migraine sphere needs to be reconsidered. “Its singular emphasis has limited our full appreciation of this disease and how we should be paying attention to the things that are important to patients,” he said.
Dr. Shapiro predicts that future clinical trials will include migraine-related stigma as a measure to guide trial enrollment.
In addition, researchers are now digging deeper to try to understand what factors are most likely to drive stigma. “We need to understand why those attitudes are held and who is more likely to hold those attitudes, and that may allow us to develop mitigating strategies to reduce those attitudes,” said Dr. Shapiro.
‘Worrisome’ findings
Commenting on the findings, Deborah Friedman, MD, professor, departments of neurology and of ophthalmology, UT Southwestern Medical Center, Dallas, said the data on stigma were “worrisome.”
Missing social occasions and lost productivity may make migraine more visible to others so perhaps may “provoke stigma or stigmatizing comments or stigmatizing attitudes,” said Dr. Friedman, who was not involved with the research.
She noted that patients with migraine might also have trouble functioning in the workplace. “They may not be able to tolerate the computer screen or smells of perfume at the office and not get accommodation for that,” she said.
In addition to external stigma, which was investigated in the study, individuals with migraine may also experience internal stigma – blaming themselves for their disease, which may make it less likely they will seek care, said Dr. Friedman.
“That’s a huge problem,” she added.
The OVERCOME study is funded by Lilly. Dr. Shapiro has been compensated by Lilly as a research consultant and as a member of the data monitoring committee for clinical trials for galcanezumab, a Lilly pharmaceutical. Dr. Friedman has reported no relevant financial relationships.
A version of this article first appeared on Medscape.com.
FROM AHS 2022
Deaths rare in tonsillectomy, but some children at more risk
It’s rare for a child to die after a tonsillectomy, but children who die are more likely to have a complex chronic condition such as cerebral palsy or Down syndrome, according to a retrospective cohort study published in JAMA.
“Among children undergoing tonsillectomy, the rate of postoperative death was 7 per 100,000 operations overall, [but] among children with complex chronic conditions, the rate of postoperative death was 117 per 100,000 operations, representing 44% of overall deaths,” write researchers at the University of Wisconsin–Madison. “These findings may inform decisionmaking for pediatric tonsillectomy.”
The rate of death in children after tonsillectomy has been uncertain, the authors write. Specific mortality rates for children at increased risk for complications, including those under 3 years old and those with sleep-disordered breathing or complex chronic conditions, have not been available.
To learn how likely children undergoing tonsillectomy are to die after their surgery, as well as which children are most at risk, lead study author M. Bruce Edmonson, MD, MPH, department of pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, and his colleagues drew data from five states, including ambulatory surgery, inpatient, and emergency department discharge data sets provided by the Healthcare Cost and Utilization Project, Agency for Healthcare Research and Quality for California, Florida, Maryland, New York, and Wisconsin.
Participants included 504,262 patients under 21 years of age whose discharge records linked their inpatient or outpatient tonsillectomy, with or without adenoidectomy, with at least 90 days of follow-up.
In a longitudinal analysis, the research team investigated postoperative death within 30 days or during a surgical stay lasting over 30 days. They calculated postoperative mortality per 100,000 operations, both overall and classified by age group, sleep-disordered breathing, and complex chronic conditions.
The 504,262 children ranged in age from 0 to 20 years and underwent a total of 505,182 tonsillectomies. Of these, 10.1% were performed in children aged under 3 years, 28.9% in children with sleep-disordered breathing, and 2.8% in those with complex chronic conditions.
The 36 linked postoperative deaths occurred between 2 and 20.5 days after surgical admission, and 19 (53%) of the deaths occurred after surgical discharge.
The unadjusted mortality rate was 7.04 (95% confidence interval, 4.97-9.98) deaths per 100,000 procedures. In multivariable models, children younger than 3 years and children with sleep-disordered breathing were not significantly more likely to die.
But children with complex chronic conditions were significantly more likely to die than were children without those conditions (117.22 vs. 3.87 deaths per 100,000 procedures, respectively).
Children with complex chronic conditions underwent only 2.8% of all tonsillectomies, but they accounted for 44% of postoperative deaths. Most deaths linked with complex chronic conditions occurred among children with neurologic, neuromuscular, congenital, or genetic disorders.
Findings can help providers advise patients and their families about tonsillectomy risks
Kavita Dedhia, MD, MSHP, attending otolaryngologist, Division of Otolaryngology, Children’s Hospital of Philadelphia, Pennsylvania, told this news organization that she was not surprised by the findings.
“This study suggests that mortality is an extremely rare complication of tonsillectomy, and that children with complex medical conditions are at highest risk,” Dr. Dedhia, who was not involved in the study, said in an email.
“Due to their underlying comorbidities, medically fragile children are considered to be at higher risk while undergoing anesthesia and surgical procedures,” she added.
Dr. Dedhia noted that nonpatient factors the study did not explore may have affected the mortality rates, including each hospital’s experience with managing children with complex medical conditions, as well as whether the hospitals were tertiary care facilities, and pediatric or adult hospitals.
She would like to know what hospital or practice characteristics may have contributed to the mortality risk and whether increased mortality in these patients is limited to tonsillectomy or is also found with other surgical procedures.
“The strength of this study is that it is large and multi-regional and that it informs providers about patient factors impacting mortality in pediatric tonsillectomy,” Dr. Dedhia said. “This study arms surgeons with data to discuss mortality risk with the families of medically complex children undergoing tonsillectomy.”
The study authors and Dr. Dedhia report no relevant financial relationships. Funding information was not provided.
A version of this article first appeared on Medscape.com.
It’s rare for a child to die after a tonsillectomy, but children who die are more likely to have a complex chronic condition such as cerebral palsy or Down syndrome, according to a retrospective cohort study published in JAMA.
“Among children undergoing tonsillectomy, the rate of postoperative death was 7 per 100,000 operations overall, [but] among children with complex chronic conditions, the rate of postoperative death was 117 per 100,000 operations, representing 44% of overall deaths,” write researchers at the University of Wisconsin–Madison. “These findings may inform decisionmaking for pediatric tonsillectomy.”
The rate of death in children after tonsillectomy has been uncertain, the authors write. Specific mortality rates for children at increased risk for complications, including those under 3 years old and those with sleep-disordered breathing or complex chronic conditions, have not been available.
To learn how likely children undergoing tonsillectomy are to die after their surgery, as well as which children are most at risk, lead study author M. Bruce Edmonson, MD, MPH, department of pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, and his colleagues drew data from five states, including ambulatory surgery, inpatient, and emergency department discharge data sets provided by the Healthcare Cost and Utilization Project, Agency for Healthcare Research and Quality for California, Florida, Maryland, New York, and Wisconsin.
Participants included 504,262 patients under 21 years of age whose discharge records linked their inpatient or outpatient tonsillectomy, with or without adenoidectomy, with at least 90 days of follow-up.
In a longitudinal analysis, the research team investigated postoperative death within 30 days or during a surgical stay lasting over 30 days. They calculated postoperative mortality per 100,000 operations, both overall and classified by age group, sleep-disordered breathing, and complex chronic conditions.
The 504,262 children ranged in age from 0 to 20 years and underwent a total of 505,182 tonsillectomies. Of these, 10.1% were performed in children aged under 3 years, 28.9% in children with sleep-disordered breathing, and 2.8% in those with complex chronic conditions.
The 36 linked postoperative deaths occurred between 2 and 20.5 days after surgical admission, and 19 (53%) of the deaths occurred after surgical discharge.
The unadjusted mortality rate was 7.04 (95% confidence interval, 4.97-9.98) deaths per 100,000 procedures. In multivariable models, children younger than 3 years and children with sleep-disordered breathing were not significantly more likely to die.
But children with complex chronic conditions were significantly more likely to die than were children without those conditions (117.22 vs. 3.87 deaths per 100,000 procedures, respectively).
Children with complex chronic conditions underwent only 2.8% of all tonsillectomies, but they accounted for 44% of postoperative deaths. Most deaths linked with complex chronic conditions occurred among children with neurologic, neuromuscular, congenital, or genetic disorders.
Findings can help providers advise patients and their families about tonsillectomy risks
Kavita Dedhia, MD, MSHP, attending otolaryngologist, Division of Otolaryngology, Children’s Hospital of Philadelphia, Pennsylvania, told this news organization that she was not surprised by the findings.
“This study suggests that mortality is an extremely rare complication of tonsillectomy, and that children with complex medical conditions are at highest risk,” Dr. Dedhia, who was not involved in the study, said in an email.
“Due to their underlying comorbidities, medically fragile children are considered to be at higher risk while undergoing anesthesia and surgical procedures,” she added.
Dr. Dedhia noted that nonpatient factors the study did not explore may have affected the mortality rates, including each hospital’s experience with managing children with complex medical conditions, as well as whether the hospitals were tertiary care facilities, and pediatric or adult hospitals.
She would like to know what hospital or practice characteristics may have contributed to the mortality risk and whether increased mortality in these patients is limited to tonsillectomy or is also found with other surgical procedures.
“The strength of this study is that it is large and multi-regional and that it informs providers about patient factors impacting mortality in pediatric tonsillectomy,” Dr. Dedhia said. “This study arms surgeons with data to discuss mortality risk with the families of medically complex children undergoing tonsillectomy.”
The study authors and Dr. Dedhia report no relevant financial relationships. Funding information was not provided.
A version of this article first appeared on Medscape.com.
It’s rare for a child to die after a tonsillectomy, but children who die are more likely to have a complex chronic condition such as cerebral palsy or Down syndrome, according to a retrospective cohort study published in JAMA.
“Among children undergoing tonsillectomy, the rate of postoperative death was 7 per 100,000 operations overall, [but] among children with complex chronic conditions, the rate of postoperative death was 117 per 100,000 operations, representing 44% of overall deaths,” write researchers at the University of Wisconsin–Madison. “These findings may inform decisionmaking for pediatric tonsillectomy.”
The rate of death in children after tonsillectomy has been uncertain, the authors write. Specific mortality rates for children at increased risk for complications, including those under 3 years old and those with sleep-disordered breathing or complex chronic conditions, have not been available.
To learn how likely children undergoing tonsillectomy are to die after their surgery, as well as which children are most at risk, lead study author M. Bruce Edmonson, MD, MPH, department of pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, and his colleagues drew data from five states, including ambulatory surgery, inpatient, and emergency department discharge data sets provided by the Healthcare Cost and Utilization Project, Agency for Healthcare Research and Quality for California, Florida, Maryland, New York, and Wisconsin.
Participants included 504,262 patients under 21 years of age whose discharge records linked their inpatient or outpatient tonsillectomy, with or without adenoidectomy, with at least 90 days of follow-up.
In a longitudinal analysis, the research team investigated postoperative death within 30 days or during a surgical stay lasting over 30 days. They calculated postoperative mortality per 100,000 operations, both overall and classified by age group, sleep-disordered breathing, and complex chronic conditions.
The 504,262 children ranged in age from 0 to 20 years and underwent a total of 505,182 tonsillectomies. Of these, 10.1% were performed in children aged under 3 years, 28.9% in children with sleep-disordered breathing, and 2.8% in those with complex chronic conditions.
The 36 linked postoperative deaths occurred between 2 and 20.5 days after surgical admission, and 19 (53%) of the deaths occurred after surgical discharge.
The unadjusted mortality rate was 7.04 (95% confidence interval, 4.97-9.98) deaths per 100,000 procedures. In multivariable models, children younger than 3 years and children with sleep-disordered breathing were not significantly more likely to die.
But children with complex chronic conditions were significantly more likely to die than were children without those conditions (117.22 vs. 3.87 deaths per 100,000 procedures, respectively).
Children with complex chronic conditions underwent only 2.8% of all tonsillectomies, but they accounted for 44% of postoperative deaths. Most deaths linked with complex chronic conditions occurred among children with neurologic, neuromuscular, congenital, or genetic disorders.
Findings can help providers advise patients and their families about tonsillectomy risks
Kavita Dedhia, MD, MSHP, attending otolaryngologist, Division of Otolaryngology, Children’s Hospital of Philadelphia, Pennsylvania, told this news organization that she was not surprised by the findings.
“This study suggests that mortality is an extremely rare complication of tonsillectomy, and that children with complex medical conditions are at highest risk,” Dr. Dedhia, who was not involved in the study, said in an email.
“Due to their underlying comorbidities, medically fragile children are considered to be at higher risk while undergoing anesthesia and surgical procedures,” she added.
Dr. Dedhia noted that nonpatient factors the study did not explore may have affected the mortality rates, including each hospital’s experience with managing children with complex medical conditions, as well as whether the hospitals were tertiary care facilities, and pediatric or adult hospitals.
She would like to know what hospital or practice characteristics may have contributed to the mortality risk and whether increased mortality in these patients is limited to tonsillectomy or is also found with other surgical procedures.
“The strength of this study is that it is large and multi-regional and that it informs providers about patient factors impacting mortality in pediatric tonsillectomy,” Dr. Dedhia said. “This study arms surgeons with data to discuss mortality risk with the families of medically complex children undergoing tonsillectomy.”
The study authors and Dr. Dedhia report no relevant financial relationships. Funding information was not provided.
A version of this article first appeared on Medscape.com.
FROM JAMA
FDA orders Juul to stop selling E-cigarettes
The marketing denial order covers all the company’s products in the United States, which means Juul must stop distributing the products and remove everything on the market. That includes the Juul device and flavor replacement pods in the tobacco and menthol flavors.
“Today’s action is further progress on the FDA’s commitment to ensuring that all e-cigarette and electronic nicotine delivery system products currently being marketed to consumers meet our public health standards,” Robert Califf, MD, the FDA commissioner, said in the announcement.
“The agency has dedicated significant resources to review products from the companies that account for most of the U.S. market,” he said. “We recognize these make up a significant part of the available products and many have played a disproportionate role in the rise in youth vaping.”
The marketing denial order covers only the commercial distribution and retail sale of Juul’s products and doesn’t restrict consumer possession or use. The FDA “cannot and will not” enforce actions against consumers, the agency said.
The order comes after a 2-year review of the company’s application seeking authorization to continue selling non–fruit-flavored products, such as menthol and tobacco. The FDA determined the application “lacked sufficient evidence regarding the toxicological profile of the products to demonstrate that marketing of the products would be appropriate for the protection of the public health.”
Some of Juul’s study findings raised concerns because of “insufficient and conflicting data,” the FDA said, including potentially harmful chemicals leaching from the Juul liquid replacement pods.
“To date, the FDA has not received clinical information to suggest an immediate hazard associated with the use of the JUUL device or JUUL pods,” the agency said. “However, the [orders] issued today reflect FDA’s determination that there is insufficient evidence to assess the potential toxicological risks of using the JUUL products.”
Juul is expected to appeal the FDA’s decision, according to The New York Times.
In recent years, the FDA has reviewed marketing applications from Juul and other e-cigarette companies as anti-tobacco groups have called for new rules to limit products that led to a surge in youth vaping during the past decade. At the same time, advocates of e-cigarettes and nicotine-delivery devices have said the products help adult smokers to quit cigarettes and other tobacco products.
Juul, in particular, has been blamed for fueling the surge in underage vaping due to fruity flavors and hip marketing, according to The Wall Street Journal. The company removed sweet and fruity flavors from shelves in 2019 and has been trying to repair its reputation by limiting its marketing and focusing on adult cigarette smokers.
In 2020, all e-cigarette manufacturers in the United States were required to submit their products for FDA review to stay on the market, the newspaper reported. The agency has been weighing the potential benefits for adult cigarette smokers against the harms for young people.
The FDA banned the sale of fruit- and mint-flavored cartridges and juice pods in 2020, but menthol and tobacco-flavored products were left on the market, according to USA Today. In September 2021, the agency also banned the sale of hundreds of thousands of vaping and e-cigarette products but didn’t rule on Juul.
Meanwhile, the FDA has cleared Reynolds American and NJOY Holdings – two of Juul’s biggest rivals – to keep tobacco-flavored products on the market. Industry experts expected Juul to receive similar clearance, the Journal reported.
Juul, which was at the top of the U.S. e-cigarette market in 2018, has moved to second place behind Reynolds’s Vuse brand, the newspaper reported. The United States represents most of the company’s revenue, though its products are also available in Canada, the United Kingdom, France, Italy, and the Philippines.
Underage vaping has fallen in the United States since federal restrictions raised the legal purchase age for tobacco products to 21 and banned the sale of sweet and fruity cartridges, according to the Journal. Juul’s popularity has also dropped among youth, with other products such as Puff Bar, Vuse, and Smok becoming more popular among e-cigarette users in high school.
In a separate decision announced this week, the FDA is also moving forward with a plan to reduce the amount of nicotine in cigarettes. The decision, which has been years in the making, is aimed at prompting millions of cigarette users to quit smoking or switch to alternatives such as e-cigarettes, as well as limit the number of users who pick up smoking at an early age.
A version of this article first appeared on WebMD.com .
The marketing denial order covers all the company’s products in the United States, which means Juul must stop distributing the products and remove everything on the market. That includes the Juul device and flavor replacement pods in the tobacco and menthol flavors.
“Today’s action is further progress on the FDA’s commitment to ensuring that all e-cigarette and electronic nicotine delivery system products currently being marketed to consumers meet our public health standards,” Robert Califf, MD, the FDA commissioner, said in the announcement.
“The agency has dedicated significant resources to review products from the companies that account for most of the U.S. market,” he said. “We recognize these make up a significant part of the available products and many have played a disproportionate role in the rise in youth vaping.”
The marketing denial order covers only the commercial distribution and retail sale of Juul’s products and doesn’t restrict consumer possession or use. The FDA “cannot and will not” enforce actions against consumers, the agency said.
The order comes after a 2-year review of the company’s application seeking authorization to continue selling non–fruit-flavored products, such as menthol and tobacco. The FDA determined the application “lacked sufficient evidence regarding the toxicological profile of the products to demonstrate that marketing of the products would be appropriate for the protection of the public health.”
Some of Juul’s study findings raised concerns because of “insufficient and conflicting data,” the FDA said, including potentially harmful chemicals leaching from the Juul liquid replacement pods.
“To date, the FDA has not received clinical information to suggest an immediate hazard associated with the use of the JUUL device or JUUL pods,” the agency said. “However, the [orders] issued today reflect FDA’s determination that there is insufficient evidence to assess the potential toxicological risks of using the JUUL products.”
Juul is expected to appeal the FDA’s decision, according to The New York Times.
In recent years, the FDA has reviewed marketing applications from Juul and other e-cigarette companies as anti-tobacco groups have called for new rules to limit products that led to a surge in youth vaping during the past decade. At the same time, advocates of e-cigarettes and nicotine-delivery devices have said the products help adult smokers to quit cigarettes and other tobacco products.
Juul, in particular, has been blamed for fueling the surge in underage vaping due to fruity flavors and hip marketing, according to The Wall Street Journal. The company removed sweet and fruity flavors from shelves in 2019 and has been trying to repair its reputation by limiting its marketing and focusing on adult cigarette smokers.
In 2020, all e-cigarette manufacturers in the United States were required to submit their products for FDA review to stay on the market, the newspaper reported. The agency has been weighing the potential benefits for adult cigarette smokers against the harms for young people.
The FDA banned the sale of fruit- and mint-flavored cartridges and juice pods in 2020, but menthol and tobacco-flavored products were left on the market, according to USA Today. In September 2021, the agency also banned the sale of hundreds of thousands of vaping and e-cigarette products but didn’t rule on Juul.
Meanwhile, the FDA has cleared Reynolds American and NJOY Holdings – two of Juul’s biggest rivals – to keep tobacco-flavored products on the market. Industry experts expected Juul to receive similar clearance, the Journal reported.
Juul, which was at the top of the U.S. e-cigarette market in 2018, has moved to second place behind Reynolds’s Vuse brand, the newspaper reported. The United States represents most of the company’s revenue, though its products are also available in Canada, the United Kingdom, France, Italy, and the Philippines.
Underage vaping has fallen in the United States since federal restrictions raised the legal purchase age for tobacco products to 21 and banned the sale of sweet and fruity cartridges, according to the Journal. Juul’s popularity has also dropped among youth, with other products such as Puff Bar, Vuse, and Smok becoming more popular among e-cigarette users in high school.
In a separate decision announced this week, the FDA is also moving forward with a plan to reduce the amount of nicotine in cigarettes. The decision, which has been years in the making, is aimed at prompting millions of cigarette users to quit smoking or switch to alternatives such as e-cigarettes, as well as limit the number of users who pick up smoking at an early age.
A version of this article first appeared on WebMD.com .
The marketing denial order covers all the company’s products in the United States, which means Juul must stop distributing the products and remove everything on the market. That includes the Juul device and flavor replacement pods in the tobacco and menthol flavors.
“Today’s action is further progress on the FDA’s commitment to ensuring that all e-cigarette and electronic nicotine delivery system products currently being marketed to consumers meet our public health standards,” Robert Califf, MD, the FDA commissioner, said in the announcement.
“The agency has dedicated significant resources to review products from the companies that account for most of the U.S. market,” he said. “We recognize these make up a significant part of the available products and many have played a disproportionate role in the rise in youth vaping.”
The marketing denial order covers only the commercial distribution and retail sale of Juul’s products and doesn’t restrict consumer possession or use. The FDA “cannot and will not” enforce actions against consumers, the agency said.
The order comes after a 2-year review of the company’s application seeking authorization to continue selling non–fruit-flavored products, such as menthol and tobacco. The FDA determined the application “lacked sufficient evidence regarding the toxicological profile of the products to demonstrate that marketing of the products would be appropriate for the protection of the public health.”
Some of Juul’s study findings raised concerns because of “insufficient and conflicting data,” the FDA said, including potentially harmful chemicals leaching from the Juul liquid replacement pods.
“To date, the FDA has not received clinical information to suggest an immediate hazard associated with the use of the JUUL device or JUUL pods,” the agency said. “However, the [orders] issued today reflect FDA’s determination that there is insufficient evidence to assess the potential toxicological risks of using the JUUL products.”
Juul is expected to appeal the FDA’s decision, according to The New York Times.
In recent years, the FDA has reviewed marketing applications from Juul and other e-cigarette companies as anti-tobacco groups have called for new rules to limit products that led to a surge in youth vaping during the past decade. At the same time, advocates of e-cigarettes and nicotine-delivery devices have said the products help adult smokers to quit cigarettes and other tobacco products.
Juul, in particular, has been blamed for fueling the surge in underage vaping due to fruity flavors and hip marketing, according to The Wall Street Journal. The company removed sweet and fruity flavors from shelves in 2019 and has been trying to repair its reputation by limiting its marketing and focusing on adult cigarette smokers.
In 2020, all e-cigarette manufacturers in the United States were required to submit their products for FDA review to stay on the market, the newspaper reported. The agency has been weighing the potential benefits for adult cigarette smokers against the harms for young people.
The FDA banned the sale of fruit- and mint-flavored cartridges and juice pods in 2020, but menthol and tobacco-flavored products were left on the market, according to USA Today. In September 2021, the agency also banned the sale of hundreds of thousands of vaping and e-cigarette products but didn’t rule on Juul.
Meanwhile, the FDA has cleared Reynolds American and NJOY Holdings – two of Juul’s biggest rivals – to keep tobacco-flavored products on the market. Industry experts expected Juul to receive similar clearance, the Journal reported.
Juul, which was at the top of the U.S. e-cigarette market in 2018, has moved to second place behind Reynolds’s Vuse brand, the newspaper reported. The United States represents most of the company’s revenue, though its products are also available in Canada, the United Kingdom, France, Italy, and the Philippines.
Underage vaping has fallen in the United States since federal restrictions raised the legal purchase age for tobacco products to 21 and banned the sale of sweet and fruity cartridges, according to the Journal. Juul’s popularity has also dropped among youth, with other products such as Puff Bar, Vuse, and Smok becoming more popular among e-cigarette users in high school.
In a separate decision announced this week, the FDA is also moving forward with a plan to reduce the amount of nicotine in cigarettes. The decision, which has been years in the making, is aimed at prompting millions of cigarette users to quit smoking or switch to alternatives such as e-cigarettes, as well as limit the number of users who pick up smoking at an early age.
A version of this article first appeared on WebMD.com .
Aging HIV patients face comorbidities and hospitalizations
Thanks to effective treatment, people with HIV are living longer. But as they age, they face higher rates of age-related comorbidities and hospitalizations, according to a recent study of hospitalized patients.
Decision-makers will need to allocate resources, train providers, and plan ways to manage chronic diseases, such as diabetes and cancer, among geriatric HIV inpatients, according to the authors.
“There will be more [HIV] patients with age-related chronic conditions at an earlier age and who will utilize or will have a unique need for [health care for] these geriatric conditions,” first author Khairul A. Siddiqi, PhD, University of Florida, Gainesville, said in an interview. “Eventually, that may increase inpatient resource utilization and costs.”
The study was published online in HIV Medicine.
Aging with HIV
Analyzing the National Inpatient Sample (NIS) of the Healthcare Cost and Utilization Project, the authors compared characteristics and comorbidities linked to hospital stays among people with HIV (HSWH) to those linked to hospital stays among people without HIV (HSWOH).
The NIS is a database of hospital records that captures 20% of discharges in the United States and covers all payers. Data in this analysis covered the years 2003-2015.
Among HSWH, patients aged 50 or older accounted for an increasing proportion over time, from fewer than 25% in 2003 to over 50% by 2015, the authors found. The subgroup aged 65-80 had risen from 2.39% to 8.63% by 2015.
The authors also studied rates of eight comorbidities, termed HIV-associated non-AIDS (HANA) conditions: cardiovascular, lung, liver, neurologic, and kidney diseases; diabetes; cancer; and bone loss.
The average number of these conditions among both HSWH and HSWOH rose over time. But this change was disproportionately high among HSWH aged 50-64 and those aged 65 and older.
Over the study period, among patients aged 65 or older, six of the eight age-related conditions the researchers studied rose disproportionately among HSWH in comparison with HSWOH; among those aged 50-64, five conditions did so.
The researchers are now building on the current study of HSWH by examining rates of resource utilization, such as MRIs and procedures, Dr. Siddiqi said.
Study limitations included a lack of data from long-term facilities, potential skewing by patients hospitalized multiple times, and the inherent limitations of administrative data.
A unique group of older people
Among people with HIV (PWH) in the United States, nearly half are aged 50 or older. By 2030, this group is expected to account for some 70% of PWH.
“We need to pay attention to what we know about aging generally. It is also important to study aging in this special population, because we don’t necessarily know a lot about that,” Amy Justice, MD, PhD, professor of medicine and of public health at Yale University, New Haven, Conn., said in an interview. Dr. Justice was not involved in the study.
The HIV epidemic has disproportionately affected people of color, men who have sex with men, and people with a history of injection drug use, Dr. Justice said.
“We don’t know about aging with [a] past history of injection drug use. We don’t even know much about aging with hepatitis C, necessarily,” she said. “So there are lots of reasons to pay some attention to this population to try to optimize their care.”
In addition, compared with their non–HIV-affected counterparts, these individuals are more susceptible to HANA comorbidities. They may experience these conditions at a younger age or more severely. Chronic inflammation and polypharmacy may be to blame, said Dr. Justice.
Given the burden of comorbidities and polypharmacy in this patient population, Dr. Siddiqi said, policy makers will need to focus on developing chronic disease management interventions for them.
However, Dr. Justice added, the risk for multimorbidity is higher among people with HIV throughout the age cycle: “It’s not like I turn 50 with HIV and all of a sudden all the wheels come off. There are ways to successfully age with HIV.”
Geriatric HIV expertise needed
Dr. Justice called the study’s analysis a useful addition to the literature and noted its implications for training.
“One of the biggest challenges with this large bolus of folks who are aging with HIV,” she said, “is to what extent should they be cared for by the people who have been caring for them – largely infectious disease docs – and to what extent should we really be transitioning their care to people with more experience with aging.”
Another key question, Dr. Justice said, relates to nursing homes and assisted-living facilities, whose staff may lack experience caring for HIV patients. Training them and hospital-based providers is crucial, in part to avoid key errors, such as missed antiretroviral doses, she said: “We need to really think about how to get non-HIV providers up to speed.”
That may begin by simply making it clear that this population is here.
“A decade ago, HIV patients used to have a lower life expectancy, so all HIV studies used to use 50 years as the cutoff point for [the] older population,” Dr. Siddiqi said. “Now we know they’re living longer.”
Added Dr. Justice: “Previously, people thought aging and HIV were not coincident findings.”
The study was funded by the Office of the Vice President for Research at the University of South Carolina. The authors and Dr. Justice disclosed no relevant financial relationships.
A version of this article first appeared on Medscape.com.
Thanks to effective treatment, people with HIV are living longer. But as they age, they face higher rates of age-related comorbidities and hospitalizations, according to a recent study of hospitalized patients.
Decision-makers will need to allocate resources, train providers, and plan ways to manage chronic diseases, such as diabetes and cancer, among geriatric HIV inpatients, according to the authors.
“There will be more [HIV] patients with age-related chronic conditions at an earlier age and who will utilize or will have a unique need for [health care for] these geriatric conditions,” first author Khairul A. Siddiqi, PhD, University of Florida, Gainesville, said in an interview. “Eventually, that may increase inpatient resource utilization and costs.”
The study was published online in HIV Medicine.
Aging with HIV
Analyzing the National Inpatient Sample (NIS) of the Healthcare Cost and Utilization Project, the authors compared characteristics and comorbidities linked to hospital stays among people with HIV (HSWH) to those linked to hospital stays among people without HIV (HSWOH).
The NIS is a database of hospital records that captures 20% of discharges in the United States and covers all payers. Data in this analysis covered the years 2003-2015.
Among HSWH, patients aged 50 or older accounted for an increasing proportion over time, from fewer than 25% in 2003 to over 50% by 2015, the authors found. The subgroup aged 65-80 had risen from 2.39% to 8.63% by 2015.
The authors also studied rates of eight comorbidities, termed HIV-associated non-AIDS (HANA) conditions: cardiovascular, lung, liver, neurologic, and kidney diseases; diabetes; cancer; and bone loss.
The average number of these conditions among both HSWH and HSWOH rose over time. But this change was disproportionately high among HSWH aged 50-64 and those aged 65 and older.
Over the study period, among patients aged 65 or older, six of the eight age-related conditions the researchers studied rose disproportionately among HSWH in comparison with HSWOH; among those aged 50-64, five conditions did so.
The researchers are now building on the current study of HSWH by examining rates of resource utilization, such as MRIs and procedures, Dr. Siddiqi said.
Study limitations included a lack of data from long-term facilities, potential skewing by patients hospitalized multiple times, and the inherent limitations of administrative data.
A unique group of older people
Among people with HIV (PWH) in the United States, nearly half are aged 50 or older. By 2030, this group is expected to account for some 70% of PWH.
“We need to pay attention to what we know about aging generally. It is also important to study aging in this special population, because we don’t necessarily know a lot about that,” Amy Justice, MD, PhD, professor of medicine and of public health at Yale University, New Haven, Conn., said in an interview. Dr. Justice was not involved in the study.
The HIV epidemic has disproportionately affected people of color, men who have sex with men, and people with a history of injection drug use, Dr. Justice said.
“We don’t know about aging with [a] past history of injection drug use. We don’t even know much about aging with hepatitis C, necessarily,” she said. “So there are lots of reasons to pay some attention to this population to try to optimize their care.”
In addition, compared with their non–HIV-affected counterparts, these individuals are more susceptible to HANA comorbidities. They may experience these conditions at a younger age or more severely. Chronic inflammation and polypharmacy may be to blame, said Dr. Justice.
Given the burden of comorbidities and polypharmacy in this patient population, Dr. Siddiqi said, policy makers will need to focus on developing chronic disease management interventions for them.
However, Dr. Justice added, the risk for multimorbidity is higher among people with HIV throughout the age cycle: “It’s not like I turn 50 with HIV and all of a sudden all the wheels come off. There are ways to successfully age with HIV.”
Geriatric HIV expertise needed
Dr. Justice called the study’s analysis a useful addition to the literature and noted its implications for training.
“One of the biggest challenges with this large bolus of folks who are aging with HIV,” she said, “is to what extent should they be cared for by the people who have been caring for them – largely infectious disease docs – and to what extent should we really be transitioning their care to people with more experience with aging.”
Another key question, Dr. Justice said, relates to nursing homes and assisted-living facilities, whose staff may lack experience caring for HIV patients. Training them and hospital-based providers is crucial, in part to avoid key errors, such as missed antiretroviral doses, she said: “We need to really think about how to get non-HIV providers up to speed.”
That may begin by simply making it clear that this population is here.
“A decade ago, HIV patients used to have a lower life expectancy, so all HIV studies used to use 50 years as the cutoff point for [the] older population,” Dr. Siddiqi said. “Now we know they’re living longer.”
Added Dr. Justice: “Previously, people thought aging and HIV were not coincident findings.”
The study was funded by the Office of the Vice President for Research at the University of South Carolina. The authors and Dr. Justice disclosed no relevant financial relationships.
A version of this article first appeared on Medscape.com.
Thanks to effective treatment, people with HIV are living longer. But as they age, they face higher rates of age-related comorbidities and hospitalizations, according to a recent study of hospitalized patients.
Decision-makers will need to allocate resources, train providers, and plan ways to manage chronic diseases, such as diabetes and cancer, among geriatric HIV inpatients, according to the authors.
“There will be more [HIV] patients with age-related chronic conditions at an earlier age and who will utilize or will have a unique need for [health care for] these geriatric conditions,” first author Khairul A. Siddiqi, PhD, University of Florida, Gainesville, said in an interview. “Eventually, that may increase inpatient resource utilization and costs.”
The study was published online in HIV Medicine.
Aging with HIV
Analyzing the National Inpatient Sample (NIS) of the Healthcare Cost and Utilization Project, the authors compared characteristics and comorbidities linked to hospital stays among people with HIV (HSWH) to those linked to hospital stays among people without HIV (HSWOH).
The NIS is a database of hospital records that captures 20% of discharges in the United States and covers all payers. Data in this analysis covered the years 2003-2015.
Among HSWH, patients aged 50 or older accounted for an increasing proportion over time, from fewer than 25% in 2003 to over 50% by 2015, the authors found. The subgroup aged 65-80 had risen from 2.39% to 8.63% by 2015.
The authors also studied rates of eight comorbidities, termed HIV-associated non-AIDS (HANA) conditions: cardiovascular, lung, liver, neurologic, and kidney diseases; diabetes; cancer; and bone loss.
The average number of these conditions among both HSWH and HSWOH rose over time. But this change was disproportionately high among HSWH aged 50-64 and those aged 65 and older.
Over the study period, among patients aged 65 or older, six of the eight age-related conditions the researchers studied rose disproportionately among HSWH in comparison with HSWOH; among those aged 50-64, five conditions did so.
The researchers are now building on the current study of HSWH by examining rates of resource utilization, such as MRIs and procedures, Dr. Siddiqi said.
Study limitations included a lack of data from long-term facilities, potential skewing by patients hospitalized multiple times, and the inherent limitations of administrative data.
A unique group of older people
Among people with HIV (PWH) in the United States, nearly half are aged 50 or older. By 2030, this group is expected to account for some 70% of PWH.
“We need to pay attention to what we know about aging generally. It is also important to study aging in this special population, because we don’t necessarily know a lot about that,” Amy Justice, MD, PhD, professor of medicine and of public health at Yale University, New Haven, Conn., said in an interview. Dr. Justice was not involved in the study.
The HIV epidemic has disproportionately affected people of color, men who have sex with men, and people with a history of injection drug use, Dr. Justice said.
“We don’t know about aging with [a] past history of injection drug use. We don’t even know much about aging with hepatitis C, necessarily,” she said. “So there are lots of reasons to pay some attention to this population to try to optimize their care.”
In addition, compared with their non–HIV-affected counterparts, these individuals are more susceptible to HANA comorbidities. They may experience these conditions at a younger age or more severely. Chronic inflammation and polypharmacy may be to blame, said Dr. Justice.
Given the burden of comorbidities and polypharmacy in this patient population, Dr. Siddiqi said, policy makers will need to focus on developing chronic disease management interventions for them.
However, Dr. Justice added, the risk for multimorbidity is higher among people with HIV throughout the age cycle: “It’s not like I turn 50 with HIV and all of a sudden all the wheels come off. There are ways to successfully age with HIV.”
Geriatric HIV expertise needed
Dr. Justice called the study’s analysis a useful addition to the literature and noted its implications for training.
“One of the biggest challenges with this large bolus of folks who are aging with HIV,” she said, “is to what extent should they be cared for by the people who have been caring for them – largely infectious disease docs – and to what extent should we really be transitioning their care to people with more experience with aging.”
Another key question, Dr. Justice said, relates to nursing homes and assisted-living facilities, whose staff may lack experience caring for HIV patients. Training them and hospital-based providers is crucial, in part to avoid key errors, such as missed antiretroviral doses, she said: “We need to really think about how to get non-HIV providers up to speed.”
That may begin by simply making it clear that this population is here.
“A decade ago, HIV patients used to have a lower life expectancy, so all HIV studies used to use 50 years as the cutoff point for [the] older population,” Dr. Siddiqi said. “Now we know they’re living longer.”
Added Dr. Justice: “Previously, people thought aging and HIV were not coincident findings.”
The study was funded by the Office of the Vice President for Research at the University of South Carolina. The authors and Dr. Justice disclosed no relevant financial relationships.
A version of this article first appeared on Medscape.com.
FROM HIV MEDICINE
Introduce allergens early, say French allergists
Although in many cases, food-allergen tolerance can be achieved with oral immunotherapy, primary prevention of food allergies remains crucial, according to the French Society of Allergology. In new recommendations that were presented at a session of the Congress of French Pediatric Societies, the academic society advocated early introduction of allergens for all children, starting at 4 months of age.
The latest prevention data from two major studies, LEAP and EAT, have prompted European and French experts to rethink their stance on food diversification. The new French proposals were recently published under the coordination of Dominique Sabouraud-Leclerc, MD, pediatrics department, Reims (France) University Hospital, on behalf of the Food Allergy Working Group of the French Society of Allergology.
For all newborns, regardless of whether they have a history of atopic or nonatopic dermatitis, food diversification is now recommended from 4 months of age instead of 6 months, as was previously recommended. If the child does not develop atopic dermatitis or develops only a mild form, peanuts, eggs, and nuts may be introduced at home.
However, if the child experiences severe atopic dermatitis, an allergy testing panel for peanuts, nuts, eggs, and cow’s milk proteins should be performed. An oral food challenge may be conducted at the allergist’s discretion.
Regarding peanuts, the working group proposed introducing a purée in the form of either a mixture of peanuts/hazelnuts/cashew nuts (1 level teaspoon five times a week; 2 g of protein/food per week) or a 100% peanut paste (1 scant teaspoon four times a week; 2 g of peanut protein/week). If the family is worried, the allergist can suggest monitoring the child in the clinic waiting room for 30 minutes after the first dose.
“We shouldn’t delay the introduction of the primary allergens anymore, regardless of whether children are at risk for a food allergy, and particularly a peanut allergy,” explained Stéphanie Lejeune, MD, pediatric pulmonologist and allergist at Lille (France) Regional University Hospital, who presented these new findings at the congress. “In fact, if we only target at-risk children, we overlook children with no family history who will nevertheless develop food allergies. The idea is to introduce everything, especially peanuts, between 4 and 6 months of age and to no longer do so gradually, one food after another, as was being done until now, beginning at 6 months and over. We must give priority to regularity over quantity.”
Although this approach is based on clinical trials, no real-life data are currently available.
LEAP and EAT studies support early introduction of peanuts
A study from 2021 summed up the risk factors for peanut allergy. About 61% of infants (4-11 months) had atopic dermatitis, 18% had a food allergy, 62% had a first-degree relative with a peanut allergy, and 11% had a confirmed peanut allergy. The risk of peanut allergy increased with age and severe eczema.
In 2015, the LEAP study, which was conducted in the United Kingdom with 640 infants aged 4-11 months who had risk factors for peanut allergy, revolutionized peanut-allergy primary prevention. Regardless of whether the children were sensitized or not, the number of children who developed a peanut allergy was systematically lower in the group that ingested the allergen in comparison with the “avoidance” group.
Additionally, the LEAP-ON study showed that protection against peanut allergy persisted for 12 months after cessation of consumption between ages 5 and 6 years among children who had consumed peanuts previously.
Early diversification in the general population was investigated in the EAT study, which involved 1303 breastfed infants. Of these infants, 24% had atopic dermatitis (median SCORAD score, 7.5). They were divided into two arms: avoidance and breast feeding until 6 months (standard introduction) or early introduction at 3 months (boiled egg, milk, peanuts, sesame, white fish, wheat, 2 g of protein twice a week). In the per-protocol analysis, there were 13 cases of peanut allergy in the standard introduction group; there were no cases in the early introduction group.
A version of this article first appeared on Medscape.com.
Although in many cases, food-allergen tolerance can be achieved with oral immunotherapy, primary prevention of food allergies remains crucial, according to the French Society of Allergology. In new recommendations that were presented at a session of the Congress of French Pediatric Societies, the academic society advocated early introduction of allergens for all children, starting at 4 months of age.
The latest prevention data from two major studies, LEAP and EAT, have prompted European and French experts to rethink their stance on food diversification. The new French proposals were recently published under the coordination of Dominique Sabouraud-Leclerc, MD, pediatrics department, Reims (France) University Hospital, on behalf of the Food Allergy Working Group of the French Society of Allergology.
For all newborns, regardless of whether they have a history of atopic or nonatopic dermatitis, food diversification is now recommended from 4 months of age instead of 6 months, as was previously recommended. If the child does not develop atopic dermatitis or develops only a mild form, peanuts, eggs, and nuts may be introduced at home.
However, if the child experiences severe atopic dermatitis, an allergy testing panel for peanuts, nuts, eggs, and cow’s milk proteins should be performed. An oral food challenge may be conducted at the allergist’s discretion.
Regarding peanuts, the working group proposed introducing a purée in the form of either a mixture of peanuts/hazelnuts/cashew nuts (1 level teaspoon five times a week; 2 g of protein/food per week) or a 100% peanut paste (1 scant teaspoon four times a week; 2 g of peanut protein/week). If the family is worried, the allergist can suggest monitoring the child in the clinic waiting room for 30 minutes after the first dose.
“We shouldn’t delay the introduction of the primary allergens anymore, regardless of whether children are at risk for a food allergy, and particularly a peanut allergy,” explained Stéphanie Lejeune, MD, pediatric pulmonologist and allergist at Lille (France) Regional University Hospital, who presented these new findings at the congress. “In fact, if we only target at-risk children, we overlook children with no family history who will nevertheless develop food allergies. The idea is to introduce everything, especially peanuts, between 4 and 6 months of age and to no longer do so gradually, one food after another, as was being done until now, beginning at 6 months and over. We must give priority to regularity over quantity.”
Although this approach is based on clinical trials, no real-life data are currently available.
LEAP and EAT studies support early introduction of peanuts
A study from 2021 summed up the risk factors for peanut allergy. About 61% of infants (4-11 months) had atopic dermatitis, 18% had a food allergy, 62% had a first-degree relative with a peanut allergy, and 11% had a confirmed peanut allergy. The risk of peanut allergy increased with age and severe eczema.
In 2015, the LEAP study, which was conducted in the United Kingdom with 640 infants aged 4-11 months who had risk factors for peanut allergy, revolutionized peanut-allergy primary prevention. Regardless of whether the children were sensitized or not, the number of children who developed a peanut allergy was systematically lower in the group that ingested the allergen in comparison with the “avoidance” group.
Additionally, the LEAP-ON study showed that protection against peanut allergy persisted for 12 months after cessation of consumption between ages 5 and 6 years among children who had consumed peanuts previously.
Early diversification in the general population was investigated in the EAT study, which involved 1303 breastfed infants. Of these infants, 24% had atopic dermatitis (median SCORAD score, 7.5). They were divided into two arms: avoidance and breast feeding until 6 months (standard introduction) or early introduction at 3 months (boiled egg, milk, peanuts, sesame, white fish, wheat, 2 g of protein twice a week). In the per-protocol analysis, there were 13 cases of peanut allergy in the standard introduction group; there were no cases in the early introduction group.
A version of this article first appeared on Medscape.com.
Although in many cases, food-allergen tolerance can be achieved with oral immunotherapy, primary prevention of food allergies remains crucial, according to the French Society of Allergology. In new recommendations that were presented at a session of the Congress of French Pediatric Societies, the academic society advocated early introduction of allergens for all children, starting at 4 months of age.
The latest prevention data from two major studies, LEAP and EAT, have prompted European and French experts to rethink their stance on food diversification. The new French proposals were recently published under the coordination of Dominique Sabouraud-Leclerc, MD, pediatrics department, Reims (France) University Hospital, on behalf of the Food Allergy Working Group of the French Society of Allergology.
For all newborns, regardless of whether they have a history of atopic or nonatopic dermatitis, food diversification is now recommended from 4 months of age instead of 6 months, as was previously recommended. If the child does not develop atopic dermatitis or develops only a mild form, peanuts, eggs, and nuts may be introduced at home.
However, if the child experiences severe atopic dermatitis, an allergy testing panel for peanuts, nuts, eggs, and cow’s milk proteins should be performed. An oral food challenge may be conducted at the allergist’s discretion.
Regarding peanuts, the working group proposed introducing a purée in the form of either a mixture of peanuts/hazelnuts/cashew nuts (1 level teaspoon five times a week; 2 g of protein/food per week) or a 100% peanut paste (1 scant teaspoon four times a week; 2 g of peanut protein/week). If the family is worried, the allergist can suggest monitoring the child in the clinic waiting room for 30 minutes after the first dose.
“We shouldn’t delay the introduction of the primary allergens anymore, regardless of whether children are at risk for a food allergy, and particularly a peanut allergy,” explained Stéphanie Lejeune, MD, pediatric pulmonologist and allergist at Lille (France) Regional University Hospital, who presented these new findings at the congress. “In fact, if we only target at-risk children, we overlook children with no family history who will nevertheless develop food allergies. The idea is to introduce everything, especially peanuts, between 4 and 6 months of age and to no longer do so gradually, one food after another, as was being done until now, beginning at 6 months and over. We must give priority to regularity over quantity.”
Although this approach is based on clinical trials, no real-life data are currently available.
LEAP and EAT studies support early introduction of peanuts
A study from 2021 summed up the risk factors for peanut allergy. About 61% of infants (4-11 months) had atopic dermatitis, 18% had a food allergy, 62% had a first-degree relative with a peanut allergy, and 11% had a confirmed peanut allergy. The risk of peanut allergy increased with age and severe eczema.
In 2015, the LEAP study, which was conducted in the United Kingdom with 640 infants aged 4-11 months who had risk factors for peanut allergy, revolutionized peanut-allergy primary prevention. Regardless of whether the children were sensitized or not, the number of children who developed a peanut allergy was systematically lower in the group that ingested the allergen in comparison with the “avoidance” group.
Additionally, the LEAP-ON study showed that protection against peanut allergy persisted for 12 months after cessation of consumption between ages 5 and 6 years among children who had consumed peanuts previously.
Early diversification in the general population was investigated in the EAT study, which involved 1303 breastfed infants. Of these infants, 24% had atopic dermatitis (median SCORAD score, 7.5). They were divided into two arms: avoidance and breast feeding until 6 months (standard introduction) or early introduction at 3 months (boiled egg, milk, peanuts, sesame, white fish, wheat, 2 g of protein twice a week). In the per-protocol analysis, there were 13 cases of peanut allergy in the standard introduction group; there were no cases in the early introduction group.
A version of this article first appeared on Medscape.com.
Study links sleep and objective, subjective cognition
CHARLOTTE, N.C. – , preliminary findings from a pilot study of objective and subjective cognitive measures have shown.
The pilot study underscored the important role of objective sleep measures to better understand discrepancies when patients’ own reports of everyday cognitive function don’t align with objective cognitive profiles, Amy Costa, MA, a graduate student in psychology at the University of Missouri-Columbia, said in reporting the results at the annual meeting of the Associated Professional Sleep Societies.
“Between our previously published paper and these new pilot results, we’re reporting evidence that suggests sleep is playing a role between the objective and subjective cognition relationship,” Ms. Costa said in an interview. “It is possible that these older adults who are sleeping poorly may be worse at understanding how well they’re doing cognitively. That’s really important for doctors. For example, if we can’t diagnose someone with mild cognitive impairment or Alzheimer’s disease or other types of dementia earlier, then we can’t intervene as quickly.”
Sleep efficiency, cognition, and patient complaints
These findings are in agreement with those Ms. Costa and colleagues recently published in the Journal of Clinical Sleep Medicine, she said.
The current pilot study included 35 older adults with an average age of 69 years who had insomnia complaints. They completed one night of home-based polysomnography – specifically with the Sleep Profiler PSG2TM – and a battery of cognitive tests. Their average sleep deficiency was 57%, “indicating potentially pretty severe insomnia,” Ms. Costa said.
“We found that sleep efficiency – that is the percentage of time spent sleeping while in bed – moderated the association between self reports and objective measures of cognitive distractibility,” Ms. Costa said in reporting the results. “In other words, our findings suggest that individuals with lower sleep efficiency who are performing the worst cognitively have the least amount of complaints. Basically, this can be thought of as that they are overestimating their cognitive performance.”
Sleep stage versus working memory and distractibility
The pilot study also focused on how the percentage of lighter-stage sleep, or N1 sleep, moderated the associations between working memory, as measured by Sternberg performance, and memory, distractibility, and blunders measured with the Cognitive Failures Questionnaire.
At the highest percentage of N1 sleep, worse working memory was associated with fewer complaints about memory, distractibility, and blunders, Ms. Costa said.
“The percentage of lighter-stage N1 sleep and sleep efficiency moderated the association between cognitive flexibility and distractibility,” Ms. Costa said. At the lowest percentage of N1 sleep, worse cognitive flexibility was associated with more distractibility, while at the highest percentage of N1 sleep worse cognitive flexibility showed a reverse effect; it was linked to less distractibility. The lowest percentage of sleep efficiency showed an association between worse cognitive flexibility and less distractibility, but the highest percentage of SE showed an association between worse cognitive flexibility and more distractibility.
“So in terms of evaluating their cognitive performance, the worse working memory was associated with more blunder complaints in individuals with the lowest percentage of N1,” she said. “So whenever individuals were spending less time in N1, they were able to better recognized their cognitive ability.”
She added, “Overall, more light and more fragmented sleep moderated the association between worse objective and less cognitive complaints, suggesting that these individuals might be overestimating their cognitive abilities.”
The findings indicate that evaluation of objective sleep should consider objectively measured N1 and sleep efficiency to better understand when subjective cognitive complaints and neurophysiological/objective cognitive profiles don’t align, she said.
Important indicators of cognitive deficits
“Specifically, for an older adult who comes into the clinic with complaints of waking up during the night, low sleep efficiency and more lighter-stage sleep might be really important indicators that they are probably not going to be the best at identifying their cognitive abilities or deficits,” she said.
Future directions for this research include collecting more data and looking at other sleep measures, such as using rapid-eye movement sleep, as potential moderators for the relationship between cognitive outcomes, evaluating sleep architecture more closely, and evaluating outcomes in a longitudinal study, Ms. Costa said.
The importance of objectively measured sleep
“Studies like this one using objectively measured sleep are important because much of the prior literature relied on self-reported sleep measures,” said Brendan P. Lucey, MD, associate professor of neurology and head of the sleep medicine section at Washington University School of Medicine in St. Louis. “This study suggests how objectively measured sleep may mediate discrepancies in objective/subjective cognitive dysfunction. Future studies need to work out if we need to add objective sleep measures when evaluating cognitive complaints in older adults.”
Dr. Lucey, who was not involved in the study, voiced one concern with the pilot study methodology the future research should address: the use of the Sleep Profiler PSG2TM to measure N1 sleep, which, as he noted, records a single-channel electroencephalogram over the forehead. “Scoring N1 sleep relies on attenuation of the alpha rhythm over the occipital region and the Sleep Profiler is not as accurate as in-lab polysomnography for this sleep stage,” he said.
The pilot study received funding from the American Academy of Sleep Medicine Foundation. Ms. Costa and her coauthors have no disclosures. Dr. Lucey disclosed relationships with Merck, Eli Lilly, Eisai, and Beacon Biosignals.
CHARLOTTE, N.C. – , preliminary findings from a pilot study of objective and subjective cognitive measures have shown.
The pilot study underscored the important role of objective sleep measures to better understand discrepancies when patients’ own reports of everyday cognitive function don’t align with objective cognitive profiles, Amy Costa, MA, a graduate student in psychology at the University of Missouri-Columbia, said in reporting the results at the annual meeting of the Associated Professional Sleep Societies.
“Between our previously published paper and these new pilot results, we’re reporting evidence that suggests sleep is playing a role between the objective and subjective cognition relationship,” Ms. Costa said in an interview. “It is possible that these older adults who are sleeping poorly may be worse at understanding how well they’re doing cognitively. That’s really important for doctors. For example, if we can’t diagnose someone with mild cognitive impairment or Alzheimer’s disease or other types of dementia earlier, then we can’t intervene as quickly.”
Sleep efficiency, cognition, and patient complaints
These findings are in agreement with those Ms. Costa and colleagues recently published in the Journal of Clinical Sleep Medicine, she said.
The current pilot study included 35 older adults with an average age of 69 years who had insomnia complaints. They completed one night of home-based polysomnography – specifically with the Sleep Profiler PSG2TM – and a battery of cognitive tests. Their average sleep deficiency was 57%, “indicating potentially pretty severe insomnia,” Ms. Costa said.
“We found that sleep efficiency – that is the percentage of time spent sleeping while in bed – moderated the association between self reports and objective measures of cognitive distractibility,” Ms. Costa said in reporting the results. “In other words, our findings suggest that individuals with lower sleep efficiency who are performing the worst cognitively have the least amount of complaints. Basically, this can be thought of as that they are overestimating their cognitive performance.”
Sleep stage versus working memory and distractibility
The pilot study also focused on how the percentage of lighter-stage sleep, or N1 sleep, moderated the associations between working memory, as measured by Sternberg performance, and memory, distractibility, and blunders measured with the Cognitive Failures Questionnaire.
At the highest percentage of N1 sleep, worse working memory was associated with fewer complaints about memory, distractibility, and blunders, Ms. Costa said.
“The percentage of lighter-stage N1 sleep and sleep efficiency moderated the association between cognitive flexibility and distractibility,” Ms. Costa said. At the lowest percentage of N1 sleep, worse cognitive flexibility was associated with more distractibility, while at the highest percentage of N1 sleep worse cognitive flexibility showed a reverse effect; it was linked to less distractibility. The lowest percentage of sleep efficiency showed an association between worse cognitive flexibility and less distractibility, but the highest percentage of SE showed an association between worse cognitive flexibility and more distractibility.
“So in terms of evaluating their cognitive performance, the worse working memory was associated with more blunder complaints in individuals with the lowest percentage of N1,” she said. “So whenever individuals were spending less time in N1, they were able to better recognized their cognitive ability.”
She added, “Overall, more light and more fragmented sleep moderated the association between worse objective and less cognitive complaints, suggesting that these individuals might be overestimating their cognitive abilities.”
The findings indicate that evaluation of objective sleep should consider objectively measured N1 and sleep efficiency to better understand when subjective cognitive complaints and neurophysiological/objective cognitive profiles don’t align, she said.
Important indicators of cognitive deficits
“Specifically, for an older adult who comes into the clinic with complaints of waking up during the night, low sleep efficiency and more lighter-stage sleep might be really important indicators that they are probably not going to be the best at identifying their cognitive abilities or deficits,” she said.
Future directions for this research include collecting more data and looking at other sleep measures, such as using rapid-eye movement sleep, as potential moderators for the relationship between cognitive outcomes, evaluating sleep architecture more closely, and evaluating outcomes in a longitudinal study, Ms. Costa said.
The importance of objectively measured sleep
“Studies like this one using objectively measured sleep are important because much of the prior literature relied on self-reported sleep measures,” said Brendan P. Lucey, MD, associate professor of neurology and head of the sleep medicine section at Washington University School of Medicine in St. Louis. “This study suggests how objectively measured sleep may mediate discrepancies in objective/subjective cognitive dysfunction. Future studies need to work out if we need to add objective sleep measures when evaluating cognitive complaints in older adults.”
Dr. Lucey, who was not involved in the study, voiced one concern with the pilot study methodology the future research should address: the use of the Sleep Profiler PSG2TM to measure N1 sleep, which, as he noted, records a single-channel electroencephalogram over the forehead. “Scoring N1 sleep relies on attenuation of the alpha rhythm over the occipital region and the Sleep Profiler is not as accurate as in-lab polysomnography for this sleep stage,” he said.
The pilot study received funding from the American Academy of Sleep Medicine Foundation. Ms. Costa and her coauthors have no disclosures. Dr. Lucey disclosed relationships with Merck, Eli Lilly, Eisai, and Beacon Biosignals.
CHARLOTTE, N.C. – , preliminary findings from a pilot study of objective and subjective cognitive measures have shown.
The pilot study underscored the important role of objective sleep measures to better understand discrepancies when patients’ own reports of everyday cognitive function don’t align with objective cognitive profiles, Amy Costa, MA, a graduate student in psychology at the University of Missouri-Columbia, said in reporting the results at the annual meeting of the Associated Professional Sleep Societies.
“Between our previously published paper and these new pilot results, we’re reporting evidence that suggests sleep is playing a role between the objective and subjective cognition relationship,” Ms. Costa said in an interview. “It is possible that these older adults who are sleeping poorly may be worse at understanding how well they’re doing cognitively. That’s really important for doctors. For example, if we can’t diagnose someone with mild cognitive impairment or Alzheimer’s disease or other types of dementia earlier, then we can’t intervene as quickly.”
Sleep efficiency, cognition, and patient complaints
These findings are in agreement with those Ms. Costa and colleagues recently published in the Journal of Clinical Sleep Medicine, she said.
The current pilot study included 35 older adults with an average age of 69 years who had insomnia complaints. They completed one night of home-based polysomnography – specifically with the Sleep Profiler PSG2TM – and a battery of cognitive tests. Their average sleep deficiency was 57%, “indicating potentially pretty severe insomnia,” Ms. Costa said.
“We found that sleep efficiency – that is the percentage of time spent sleeping while in bed – moderated the association between self reports and objective measures of cognitive distractibility,” Ms. Costa said in reporting the results. “In other words, our findings suggest that individuals with lower sleep efficiency who are performing the worst cognitively have the least amount of complaints. Basically, this can be thought of as that they are overestimating their cognitive performance.”
Sleep stage versus working memory and distractibility
The pilot study also focused on how the percentage of lighter-stage sleep, or N1 sleep, moderated the associations between working memory, as measured by Sternberg performance, and memory, distractibility, and blunders measured with the Cognitive Failures Questionnaire.
At the highest percentage of N1 sleep, worse working memory was associated with fewer complaints about memory, distractibility, and blunders, Ms. Costa said.
“The percentage of lighter-stage N1 sleep and sleep efficiency moderated the association between cognitive flexibility and distractibility,” Ms. Costa said. At the lowest percentage of N1 sleep, worse cognitive flexibility was associated with more distractibility, while at the highest percentage of N1 sleep worse cognitive flexibility showed a reverse effect; it was linked to less distractibility. The lowest percentage of sleep efficiency showed an association between worse cognitive flexibility and less distractibility, but the highest percentage of SE showed an association between worse cognitive flexibility and more distractibility.
“So in terms of evaluating their cognitive performance, the worse working memory was associated with more blunder complaints in individuals with the lowest percentage of N1,” she said. “So whenever individuals were spending less time in N1, they were able to better recognized their cognitive ability.”
She added, “Overall, more light and more fragmented sleep moderated the association between worse objective and less cognitive complaints, suggesting that these individuals might be overestimating their cognitive abilities.”
The findings indicate that evaluation of objective sleep should consider objectively measured N1 and sleep efficiency to better understand when subjective cognitive complaints and neurophysiological/objective cognitive profiles don’t align, she said.
Important indicators of cognitive deficits
“Specifically, for an older adult who comes into the clinic with complaints of waking up during the night, low sleep efficiency and more lighter-stage sleep might be really important indicators that they are probably not going to be the best at identifying their cognitive abilities or deficits,” she said.
Future directions for this research include collecting more data and looking at other sleep measures, such as using rapid-eye movement sleep, as potential moderators for the relationship between cognitive outcomes, evaluating sleep architecture more closely, and evaluating outcomes in a longitudinal study, Ms. Costa said.
The importance of objectively measured sleep
“Studies like this one using objectively measured sleep are important because much of the prior literature relied on self-reported sleep measures,” said Brendan P. Lucey, MD, associate professor of neurology and head of the sleep medicine section at Washington University School of Medicine in St. Louis. “This study suggests how objectively measured sleep may mediate discrepancies in objective/subjective cognitive dysfunction. Future studies need to work out if we need to add objective sleep measures when evaluating cognitive complaints in older adults.”
Dr. Lucey, who was not involved in the study, voiced one concern with the pilot study methodology the future research should address: the use of the Sleep Profiler PSG2TM to measure N1 sleep, which, as he noted, records a single-channel electroencephalogram over the forehead. “Scoring N1 sleep relies on attenuation of the alpha rhythm over the occipital region and the Sleep Profiler is not as accurate as in-lab polysomnography for this sleep stage,” he said.
The pilot study received funding from the American Academy of Sleep Medicine Foundation. Ms. Costa and her coauthors have no disclosures. Dr. Lucey disclosed relationships with Merck, Eli Lilly, Eisai, and Beacon Biosignals.
AT SLEEP 2022
Tofersen linked to slow, positive effects in ALS
caused by superoxide dismutase 1 (SOD1) gene mutations.
The 1-year results, presented at the European Network for the Cure of Amyotrophic Lateral Sclerosis (ENCALS) 2022 meeting, show a deceleration in functional decline that is similar, but “more pronounced” than the previously reported 6-month results, which were not statistically significant, said lead investigator Timothy Miller, MD, PhD, professor of neurology and director of the ALS Center, Washington University, St. Louis.
“What I thought we saw in the first data cut is confirmed by what we saw in the longer data,” he said in an interview. “There were trends [showing] those treated with tofersen did a bit better, but it was hard to be sure. It was hard to be confident in what we were seeing at that early time point.”
Now, with 6 more months of data, Dr. Miller says he is confident that tofersen is slowing down the neurodegenerative disease process. “I see results that I’m encouraged by,” he said. “As a clinician who treats people with ALS with this mutation I would want this drug to be available to people that I see in my clinic.”
One-year VALOR study results
The primary efficacy objective of the VALOR study was to show the 28-week impact of 100 mg tofersen (three doses given about 2 weeks apart, then five doses given every 4 weeks), versus placebo, on function, measured on the Amyotrophic Lateral Sclerosis Functional Rating Scale–Revised (ALSFRS-R). The open-label extension switched placebo-treated patients to tofersen (delayed-start group) and continued to compare them with the early-start group up to 1 year. This open-label extension phase included 49 patients who had been on early-start tofersen and 18 patients in the delayed-start group.
For the primary endpoint, change from baseline in 48-point ALSFRS-R score, there was a statistically significant benefit for the early-start patients with these patients scoring 3.5 points higher than the delayed-start group (P = .0272, 95% confidence interval [CI], 0.4-6.7). This means that both groups declined in function, which is expected in ALS, but the early-start group declined more slowly.
There was also a benefit associated with early-start tofersen for a number of secondary endpoints, including change from baseline in total SOD1 cerebrospinal fluid concentration (CSF SOD1), plasma neurofilament light chain (NfL) levels, and respiratory function.
“This drug targets the MRNA of SOD1, so it lowers the MRNA and then the SOD1 protein falls,” explained Dr. Miller, adding that these levels dropped 21% in the delayed start group, and 33% in the early-start group. “I think the data pretty clearly show that [tofersen] does what it is supposed to do, and that is the first step.”
Neurofilament light chain, a marker of neurodegeneration, also dropped by 41% in the delayed-start group, and 51% in the early-start group.
Respiratory function, as measured by percent predicted slow vital capacity (SVC), also declined 9.2% more slowly in the early- versus delayed-start group (P = .0159).
Finally, muscle strength, as measured by handheld dynamometry (HHD) score, declined more slowly in the early-start group compared with the late-start group, with an adjusted mean difference in score of 2.8 (P = 0.0186).
Dr. Miller said that the data show that it takes time for tofersen to impact clinical function, but there are signs of benefit before that. “I think what you see is that just starting on the drug, the first thing that happens is SOD1 goes down, the next thing is that neurofilament decreases, but clinical function is not yet changing. It takes time. What I see in these data is that it takes time for us to see that effect on clinical function.”
The bigger picture
While acknowledging that tofersen acts on a genetic mutation found in only about 2% of ALS, Dr. Miller said the study findings carry significance for the wider ALS patient population.
“Assuming we agree there is a clinical effect here, assuming we agree that there is real stabilization of clinical function, I think if we agree on that point then we know that ALS is now a treatable disorder. And that’s a really important point. I’m not sure that we knew that before,” he said. “Yes, there are FDA-approved medications that slow down ALS a bit, but they don’t stabilize it, and if we get the target correct – and we have the correct genetic target here – there can be a substantial influence on slowing down the disease, so that’s one thing to learn for the whole ALS community.”
What lies ahead?
Asked to comment on the study, Richard Bedlack, MD, PhD, who was not involved in the research, said the findings are important and show “clinically meaningful” results. “Based on the new benefit-to-burden ratio, I believe most of my patients with SOD1 mutations will want to try this drug. I would like to be able to offer it to them. But I am curious to see what the FDA will do with these data,” said Dr. Bedlack, professor of neurology at Duke University in Durham, N.C., and director of the Duke ALS Clinic.
“Sometimes that open-label extension gives us time to see differences between patients who initially got drug and those who initially got placebo. That seems to be the case with tofersen here, and it was also the case with AMX0035 [Amylyx Pharmaceuticals Inc.], which did not show a survival benefit in the first 6 months but did in the open-label extension.” A recent FDA advisory board panel concluded there was insufficient evidence of benefit for AMX0035, he noted. “I wonder if the same concern will be raised here, necessitating confirmation in another trial. I hope not, but only time will tell.”
Dr. Miller added that these results “highlight how difficult ALS drug development still is. Among the many uncertainties in setting up a trial (targets, doses, inclusion criteria, outcomes), we still do not know how long we need to treat patients in order to see statistically significant changes in the clinical measures we use (ALSFRS-R, respiratory function, strength, survival, etc.). Most American studies are 6 months long and most European studies are 12 months long. Longer studies may be more likely to show benefits on certain measures (e.g., survival), but they cost more, they are challenged by dropouts as the disease progresses, and the idea of randomizing someone to a placebo for a whole year is psychologically difficult for patients, families, and many clinicians (myself included). So, we are seeing more studies like this one where the first 6 months are randomized, blinded, and placebo controlled, and then there is an open-label extension that lasts many months more.”
The study was sponsored by Biogen. Writing and editorial support was provided by Excel Scientific Solutions. Tofersen was discovered by Ionis Pharmaceuticals Inc. Dr. Miller disclosed ties with Biogen, Ionis Pharmaceuticals Inc., Cytokinetics, C2N, Disarm Therapeutics, and UCB Pharma. Dr. Bedlack disclosed ties with Biogen.
caused by superoxide dismutase 1 (SOD1) gene mutations.
The 1-year results, presented at the European Network for the Cure of Amyotrophic Lateral Sclerosis (ENCALS) 2022 meeting, show a deceleration in functional decline that is similar, but “more pronounced” than the previously reported 6-month results, which were not statistically significant, said lead investigator Timothy Miller, MD, PhD, professor of neurology and director of the ALS Center, Washington University, St. Louis.
“What I thought we saw in the first data cut is confirmed by what we saw in the longer data,” he said in an interview. “There were trends [showing] those treated with tofersen did a bit better, but it was hard to be sure. It was hard to be confident in what we were seeing at that early time point.”
Now, with 6 more months of data, Dr. Miller says he is confident that tofersen is slowing down the neurodegenerative disease process. “I see results that I’m encouraged by,” he said. “As a clinician who treats people with ALS with this mutation I would want this drug to be available to people that I see in my clinic.”
One-year VALOR study results
The primary efficacy objective of the VALOR study was to show the 28-week impact of 100 mg tofersen (three doses given about 2 weeks apart, then five doses given every 4 weeks), versus placebo, on function, measured on the Amyotrophic Lateral Sclerosis Functional Rating Scale–Revised (ALSFRS-R). The open-label extension switched placebo-treated patients to tofersen (delayed-start group) and continued to compare them with the early-start group up to 1 year. This open-label extension phase included 49 patients who had been on early-start tofersen and 18 patients in the delayed-start group.
For the primary endpoint, change from baseline in 48-point ALSFRS-R score, there was a statistically significant benefit for the early-start patients with these patients scoring 3.5 points higher than the delayed-start group (P = .0272, 95% confidence interval [CI], 0.4-6.7). This means that both groups declined in function, which is expected in ALS, but the early-start group declined more slowly.
There was also a benefit associated with early-start tofersen for a number of secondary endpoints, including change from baseline in total SOD1 cerebrospinal fluid concentration (CSF SOD1), plasma neurofilament light chain (NfL) levels, and respiratory function.
“This drug targets the MRNA of SOD1, so it lowers the MRNA and then the SOD1 protein falls,” explained Dr. Miller, adding that these levels dropped 21% in the delayed start group, and 33% in the early-start group. “I think the data pretty clearly show that [tofersen] does what it is supposed to do, and that is the first step.”
Neurofilament light chain, a marker of neurodegeneration, also dropped by 41% in the delayed-start group, and 51% in the early-start group.
Respiratory function, as measured by percent predicted slow vital capacity (SVC), also declined 9.2% more slowly in the early- versus delayed-start group (P = .0159).
Finally, muscle strength, as measured by handheld dynamometry (HHD) score, declined more slowly in the early-start group compared with the late-start group, with an adjusted mean difference in score of 2.8 (P = 0.0186).
Dr. Miller said that the data show that it takes time for tofersen to impact clinical function, but there are signs of benefit before that. “I think what you see is that just starting on the drug, the first thing that happens is SOD1 goes down, the next thing is that neurofilament decreases, but clinical function is not yet changing. It takes time. What I see in these data is that it takes time for us to see that effect on clinical function.”
The bigger picture
While acknowledging that tofersen acts on a genetic mutation found in only about 2% of ALS, Dr. Miller said the study findings carry significance for the wider ALS patient population.
“Assuming we agree there is a clinical effect here, assuming we agree that there is real stabilization of clinical function, I think if we agree on that point then we know that ALS is now a treatable disorder. And that’s a really important point. I’m not sure that we knew that before,” he said. “Yes, there are FDA-approved medications that slow down ALS a bit, but they don’t stabilize it, and if we get the target correct – and we have the correct genetic target here – there can be a substantial influence on slowing down the disease, so that’s one thing to learn for the whole ALS community.”
What lies ahead?
Asked to comment on the study, Richard Bedlack, MD, PhD, who was not involved in the research, said the findings are important and show “clinically meaningful” results. “Based on the new benefit-to-burden ratio, I believe most of my patients with SOD1 mutations will want to try this drug. I would like to be able to offer it to them. But I am curious to see what the FDA will do with these data,” said Dr. Bedlack, professor of neurology at Duke University in Durham, N.C., and director of the Duke ALS Clinic.
“Sometimes that open-label extension gives us time to see differences between patients who initially got drug and those who initially got placebo. That seems to be the case with tofersen here, and it was also the case with AMX0035 [Amylyx Pharmaceuticals Inc.], which did not show a survival benefit in the first 6 months but did in the open-label extension.” A recent FDA advisory board panel concluded there was insufficient evidence of benefit for AMX0035, he noted. “I wonder if the same concern will be raised here, necessitating confirmation in another trial. I hope not, but only time will tell.”
Dr. Miller added that these results “highlight how difficult ALS drug development still is. Among the many uncertainties in setting up a trial (targets, doses, inclusion criteria, outcomes), we still do not know how long we need to treat patients in order to see statistically significant changes in the clinical measures we use (ALSFRS-R, respiratory function, strength, survival, etc.). Most American studies are 6 months long and most European studies are 12 months long. Longer studies may be more likely to show benefits on certain measures (e.g., survival), but they cost more, they are challenged by dropouts as the disease progresses, and the idea of randomizing someone to a placebo for a whole year is psychologically difficult for patients, families, and many clinicians (myself included). So, we are seeing more studies like this one where the first 6 months are randomized, blinded, and placebo controlled, and then there is an open-label extension that lasts many months more.”
The study was sponsored by Biogen. Writing and editorial support was provided by Excel Scientific Solutions. Tofersen was discovered by Ionis Pharmaceuticals Inc. Dr. Miller disclosed ties with Biogen, Ionis Pharmaceuticals Inc., Cytokinetics, C2N, Disarm Therapeutics, and UCB Pharma. Dr. Bedlack disclosed ties with Biogen.
caused by superoxide dismutase 1 (SOD1) gene mutations.
The 1-year results, presented at the European Network for the Cure of Amyotrophic Lateral Sclerosis (ENCALS) 2022 meeting, show a deceleration in functional decline that is similar, but “more pronounced” than the previously reported 6-month results, which were not statistically significant, said lead investigator Timothy Miller, MD, PhD, professor of neurology and director of the ALS Center, Washington University, St. Louis.
“What I thought we saw in the first data cut is confirmed by what we saw in the longer data,” he said in an interview. “There were trends [showing] those treated with tofersen did a bit better, but it was hard to be sure. It was hard to be confident in what we were seeing at that early time point.”
Now, with 6 more months of data, Dr. Miller says he is confident that tofersen is slowing down the neurodegenerative disease process. “I see results that I’m encouraged by,” he said. “As a clinician who treats people with ALS with this mutation I would want this drug to be available to people that I see in my clinic.”
One-year VALOR study results
The primary efficacy objective of the VALOR study was to show the 28-week impact of 100 mg tofersen (three doses given about 2 weeks apart, then five doses given every 4 weeks), versus placebo, on function, measured on the Amyotrophic Lateral Sclerosis Functional Rating Scale–Revised (ALSFRS-R). The open-label extension switched placebo-treated patients to tofersen (delayed-start group) and continued to compare them with the early-start group up to 1 year. This open-label extension phase included 49 patients who had been on early-start tofersen and 18 patients in the delayed-start group.
For the primary endpoint, change from baseline in 48-point ALSFRS-R score, there was a statistically significant benefit for the early-start patients with these patients scoring 3.5 points higher than the delayed-start group (P = .0272, 95% confidence interval [CI], 0.4-6.7). This means that both groups declined in function, which is expected in ALS, but the early-start group declined more slowly.
There was also a benefit associated with early-start tofersen for a number of secondary endpoints, including change from baseline in total SOD1 cerebrospinal fluid concentration (CSF SOD1), plasma neurofilament light chain (NfL) levels, and respiratory function.
“This drug targets the MRNA of SOD1, so it lowers the MRNA and then the SOD1 protein falls,” explained Dr. Miller, adding that these levels dropped 21% in the delayed start group, and 33% in the early-start group. “I think the data pretty clearly show that [tofersen] does what it is supposed to do, and that is the first step.”
Neurofilament light chain, a marker of neurodegeneration, also dropped by 41% in the delayed-start group, and 51% in the early-start group.
Respiratory function, as measured by percent predicted slow vital capacity (SVC), also declined 9.2% more slowly in the early- versus delayed-start group (P = .0159).
Finally, muscle strength, as measured by handheld dynamometry (HHD) score, declined more slowly in the early-start group compared with the late-start group, with an adjusted mean difference in score of 2.8 (P = 0.0186).
Dr. Miller said that the data show that it takes time for tofersen to impact clinical function, but there are signs of benefit before that. “I think what you see is that just starting on the drug, the first thing that happens is SOD1 goes down, the next thing is that neurofilament decreases, but clinical function is not yet changing. It takes time. What I see in these data is that it takes time for us to see that effect on clinical function.”
The bigger picture
While acknowledging that tofersen acts on a genetic mutation found in only about 2% of ALS, Dr. Miller said the study findings carry significance for the wider ALS patient population.
“Assuming we agree there is a clinical effect here, assuming we agree that there is real stabilization of clinical function, I think if we agree on that point then we know that ALS is now a treatable disorder. And that’s a really important point. I’m not sure that we knew that before,” he said. “Yes, there are FDA-approved medications that slow down ALS a bit, but they don’t stabilize it, and if we get the target correct – and we have the correct genetic target here – there can be a substantial influence on slowing down the disease, so that’s one thing to learn for the whole ALS community.”
What lies ahead?
Asked to comment on the study, Richard Bedlack, MD, PhD, who was not involved in the research, said the findings are important and show “clinically meaningful” results. “Based on the new benefit-to-burden ratio, I believe most of my patients with SOD1 mutations will want to try this drug. I would like to be able to offer it to them. But I am curious to see what the FDA will do with these data,” said Dr. Bedlack, professor of neurology at Duke University in Durham, N.C., and director of the Duke ALS Clinic.
“Sometimes that open-label extension gives us time to see differences between patients who initially got drug and those who initially got placebo. That seems to be the case with tofersen here, and it was also the case with AMX0035 [Amylyx Pharmaceuticals Inc.], which did not show a survival benefit in the first 6 months but did in the open-label extension.” A recent FDA advisory board panel concluded there was insufficient evidence of benefit for AMX0035, he noted. “I wonder if the same concern will be raised here, necessitating confirmation in another trial. I hope not, but only time will tell.”
Dr. Miller added that these results “highlight how difficult ALS drug development still is. Among the many uncertainties in setting up a trial (targets, doses, inclusion criteria, outcomes), we still do not know how long we need to treat patients in order to see statistically significant changes in the clinical measures we use (ALSFRS-R, respiratory function, strength, survival, etc.). Most American studies are 6 months long and most European studies are 12 months long. Longer studies may be more likely to show benefits on certain measures (e.g., survival), but they cost more, they are challenged by dropouts as the disease progresses, and the idea of randomizing someone to a placebo for a whole year is psychologically difficult for patients, families, and many clinicians (myself included). So, we are seeing more studies like this one where the first 6 months are randomized, blinded, and placebo controlled, and then there is an open-label extension that lasts many months more.”
The study was sponsored by Biogen. Writing and editorial support was provided by Excel Scientific Solutions. Tofersen was discovered by Ionis Pharmaceuticals Inc. Dr. Miller disclosed ties with Biogen, Ionis Pharmaceuticals Inc., Cytokinetics, C2N, Disarm Therapeutics, and UCB Pharma. Dr. Bedlack disclosed ties with Biogen.
FROM THE ENCALS MEETING 2022