Patient safety vs. public health: The ethylene oxide dilemma

Article Type
Changed
Wed, 08/16/2023 - 17:16

Ethylene oxide is a compound used to sterilize more than 20 billion devices sold in the U.S. every year. Although this sterilization process helps keep medical devices – and patients – safe, the odorless, flammable gas may also be harming people who live near sterilization plants and who may inhale the compound, which has been linked to an elevated risk of cancer.

Regulatory agencies are currently feuding over the best way to address the dilemma: preserving patient safety versus protecting public health. Lawmakers are weighing in on the matter, which has been the source of multiple civil lawsuits filed by individuals who say they have suffered health problems as a result of exposure to ethylene oxide.

The Environmental Protection Agency and the U.S. Food and Drug Administration agree that use of the compound should be limited, but they are at odds about how quickly limits should be put in place, according to Axios.

A new commercial standard for ethylene oxide proposed by the EPA in April would impose stricter emission restrictions for sterilization facilities and chemical plants – a move that would cut ethylene oxide emissions by 80%, the EPA estimates.

While the FDA says it “shares concerns about the release of ethylene oxide at unsafe levels into the environment,” the agency cautions that moving too fast to cut emissions would disrupt the medical supply chain, which is already experiencing turbulence. The U.S. has been facing the worst drug supply shortages in a decade in addition to severe medical device shortages.

Currently, other methods of sterilization cannot replace the use of ethylene oxide for many devices. Ethylene oxide is used to sterilize about half of all medical devices in the U.S., the FDA says. Given the country’s reliance on this compound for sterilization, the FDA says it is “equally concerned about the potential impact of shortages of sterilized medical devices that would result from disruptions in commercial sterilizer facility operations.”

In 2019, Illinois temporarily closed a sterilization facility over concern regarding ethylene oxide emissions. The closure caused a shortage of a pediatric breathing tube.

Some lawmakers agree that an Interior-Environment bill would require FDA certification that any action by the EPA would not cause a medical device shortage.

The FDA has been working to identify safe alternatives to ethylene oxide for sterilizing medical supplies as well as strategies to reduce emissions of ethylene oxide by capturing the gas or by turning it into a harmless byproduct. In 2019, the FDA launched a pilot program to incentivize companies to develop new sterilization technologies.

“The FDA remains focused in our commitment to encourage novel ways to sterilize medical devices while reducing adverse impacts on the environment and public health and developing solutions to avoid potential shortages of devices that the American public relies upon,” the agency said.

A version of this article first appeared on Medscape.com.

Publications
Topics
Sections

Ethylene oxide is a compound used to sterilize more than 20 billion devices sold in the U.S. every year. Although this sterilization process helps keep medical devices – and patients – safe, the odorless, flammable gas may also be harming people who live near sterilization plants and who may inhale the compound, which has been linked to an elevated risk of cancer.

Regulatory agencies are currently feuding over the best way to address the dilemma: preserving patient safety versus protecting public health. Lawmakers are weighing in on the matter, which has been the source of multiple civil lawsuits filed by individuals who say they have suffered health problems as a result of exposure to ethylene oxide.

The Environmental Protection Agency and the U.S. Food and Drug Administration agree that use of the compound should be limited, but they are at odds about how quickly limits should be put in place, according to Axios.

A new commercial standard for ethylene oxide proposed by the EPA in April would impose stricter emission restrictions for sterilization facilities and chemical plants – a move that would cut ethylene oxide emissions by 80%, the EPA estimates.

While the FDA says it “shares concerns about the release of ethylene oxide at unsafe levels into the environment,” the agency cautions that moving too fast to cut emissions would disrupt the medical supply chain, which is already experiencing turbulence. The U.S. has been facing the worst drug supply shortages in a decade in addition to severe medical device shortages.

Currently, other methods of sterilization cannot replace the use of ethylene oxide for many devices. Ethylene oxide is used to sterilize about half of all medical devices in the U.S., the FDA says. Given the country’s reliance on this compound for sterilization, the FDA says it is “equally concerned about the potential impact of shortages of sterilized medical devices that would result from disruptions in commercial sterilizer facility operations.”

In 2019, Illinois temporarily closed a sterilization facility over concern regarding ethylene oxide emissions. The closure caused a shortage of a pediatric breathing tube.

Some lawmakers agree that an Interior-Environment bill would require FDA certification that any action by the EPA would not cause a medical device shortage.

The FDA has been working to identify safe alternatives to ethylene oxide for sterilizing medical supplies as well as strategies to reduce emissions of ethylene oxide by capturing the gas or by turning it into a harmless byproduct. In 2019, the FDA launched a pilot program to incentivize companies to develop new sterilization technologies.

“The FDA remains focused in our commitment to encourage novel ways to sterilize medical devices while reducing adverse impacts on the environment and public health and developing solutions to avoid potential shortages of devices that the American public relies upon,” the agency said.

A version of this article first appeared on Medscape.com.

Ethylene oxide is a compound used to sterilize more than 20 billion devices sold in the U.S. every year. Although this sterilization process helps keep medical devices – and patients – safe, the odorless, flammable gas may also be harming people who live near sterilization plants and who may inhale the compound, which has been linked to an elevated risk of cancer.

Regulatory agencies are currently feuding over the best way to address the dilemma: preserving patient safety versus protecting public health. Lawmakers are weighing in on the matter, which has been the source of multiple civil lawsuits filed by individuals who say they have suffered health problems as a result of exposure to ethylene oxide.

The Environmental Protection Agency and the U.S. Food and Drug Administration agree that use of the compound should be limited, but they are at odds about how quickly limits should be put in place, according to Axios.

A new commercial standard for ethylene oxide proposed by the EPA in April would impose stricter emission restrictions for sterilization facilities and chemical plants – a move that would cut ethylene oxide emissions by 80%, the EPA estimates.

While the FDA says it “shares concerns about the release of ethylene oxide at unsafe levels into the environment,” the agency cautions that moving too fast to cut emissions would disrupt the medical supply chain, which is already experiencing turbulence. The U.S. has been facing the worst drug supply shortages in a decade in addition to severe medical device shortages.

Currently, other methods of sterilization cannot replace the use of ethylene oxide for many devices. Ethylene oxide is used to sterilize about half of all medical devices in the U.S., the FDA says. Given the country’s reliance on this compound for sterilization, the FDA says it is “equally concerned about the potential impact of shortages of sterilized medical devices that would result from disruptions in commercial sterilizer facility operations.”

In 2019, Illinois temporarily closed a sterilization facility over concern regarding ethylene oxide emissions. The closure caused a shortage of a pediatric breathing tube.

Some lawmakers agree that an Interior-Environment bill would require FDA certification that any action by the EPA would not cause a medical device shortage.

The FDA has been working to identify safe alternatives to ethylene oxide for sterilizing medical supplies as well as strategies to reduce emissions of ethylene oxide by capturing the gas or by turning it into a harmless byproduct. In 2019, the FDA launched a pilot program to incentivize companies to develop new sterilization technologies.

“The FDA remains focused in our commitment to encourage novel ways to sterilize medical devices while reducing adverse impacts on the environment and public health and developing solutions to avoid potential shortages of devices that the American public relies upon,” the agency said.

A version of this article first appeared on Medscape.com.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Alcohol consumption may not influence breast cancer prognosis, study

Article Type
Changed
Wed, 08/16/2023 - 17:16

Drinking alcohol around the time of a breast cancer diagnosis may not have effects on prognosis that is mediated by body mass index (BMI), an analysis of data from a prospective cohort study suggests.

Kaiser Permanente
Dr. Marilyn Kwan

The study appears to show that drinking up to one serving of alcohol daily, including wine, beer, and liquor, was not associated with any specific outcomes after breast cancer diagnosis. The authors say these findings could have implications for developing more specific guidelines on alcohol use as it relates to the prevention of death and recurrence for cancer survivors.

Among 3,659 women followed for a mean of 11.2 years after a breast cancer diagnosis, overall alcohol consumption in the months before and up to 6 months after diagnosis was not associated with recurrence or mortality after adjusting for numerous factors such as age at diagnosis, cancer stage, socioeconomic details, smoking history, and preexisting conditions.

However, women with obesity (body mass index of 30 kg/m2 or greater) had a lower risk of mortality with increasing alcohol consumption for occasional drinking of 2 or more alcohol servings per week (hazard ratio, 0.71), and regular drinking of at least one alcohol serving daily (HR, 0.77), in a dose-response manner, Marilyn L. Kwan, PhD, and colleagues found.

Dr. Kwan is a senior research scientist at Kaiser Permanente Northern California Division of Research, Oakland.

Women with BMI less than 30 kg/m2 did not have a higher risk of mortality but a nonsignificant increase in the risk of recurrence was observed for those who consumed alcohol occasionally (HR, 1.29) and regularly (HR, 1.19), the investigators reported.

The findings were published online in Cancer.

Women included in the current study were participants in the Pathways Study and were diagnosed with stage I-IV breast cancer between 2003 and 2015. During follow-up, 524 recurrences and 834 deaths occurred, including 369 breast cancer-specific deaths, 314 cardiovascular disease-specific deaths, and 151 deaths from other health problems.

Alcohol consumption was assessed for the 6 months prior to cohort entry, which occurred at an average of about 2 months after diagnosis, as well as 6 months later – at an average of about 8 months after diagnosis – using a food-frequency questionnaire.

Compared with nondrinkers (36.9%), drinkers were more likely younger, more educated, and current or past smokers, the investigators noted.

“This profile appears counterintuitive yet might reflect a healthier lifestyle contributing to better overall survival. Furthermore, higher levels of alcohol consumption could lead to improvement in insulin sensitivity and reduction in insulin-like growth factor-1,” they speculated, noting that reduced fasting insulin concentrations and lower insulin-like growth factor-1 levels are linked with a decreased risk of type 2 diabetes, cardiovascular disease, and cancer.

“Many women with a history of breast cancer are interested in how to improve their prognosis and survival by making lifestyle changes after diagnosis,” they wrote, explaining the rationale for the study. “Current cancer prevention guidelines recommend avoiding alcohol intake or limiting consumption to no more than one drink per day for women. However, no specific guideline exists for cancer survivors other than following the cancer prevention guidelines to reduce the risk of a second cancer.”

High-quality studies on the impact of alcohol consumption on breast cancer prognosis are lacking, they added.

“Given that consuming alcohol is a potentially modifiable lifestyle factor after breast cancer diagnosis, further confirmation is warranted in other large prospective studies of breast cancer survivors with detailed exposure assessment and focus on body size,” they concluded.

The group is the first to report this finding in obese women, and they “strongly believe more research is needed to see if the same association is seen in other studies,” Dr. Kwan told this news organization.

“After a cancer diagnosis, many patients are motivated to make lifestyle changes,” she said. “That often includes adding exercise to their daily routine and eating a healthier diet. Our study findings suggest that doctors can tell patients that having up to a glass of alcohol a day is not likely to increase their risk of a breast cancer recurrence.”

This study was funded by the National Cancer Institute. The authors reported having no disclosures.
 

Publications
Topics
Sections

Drinking alcohol around the time of a breast cancer diagnosis may not have effects on prognosis that is mediated by body mass index (BMI), an analysis of data from a prospective cohort study suggests.

Kaiser Permanente
Dr. Marilyn Kwan

The study appears to show that drinking up to one serving of alcohol daily, including wine, beer, and liquor, was not associated with any specific outcomes after breast cancer diagnosis. The authors say these findings could have implications for developing more specific guidelines on alcohol use as it relates to the prevention of death and recurrence for cancer survivors.

Among 3,659 women followed for a mean of 11.2 years after a breast cancer diagnosis, overall alcohol consumption in the months before and up to 6 months after diagnosis was not associated with recurrence or mortality after adjusting for numerous factors such as age at diagnosis, cancer stage, socioeconomic details, smoking history, and preexisting conditions.

However, women with obesity (body mass index of 30 kg/m2 or greater) had a lower risk of mortality with increasing alcohol consumption for occasional drinking of 2 or more alcohol servings per week (hazard ratio, 0.71), and regular drinking of at least one alcohol serving daily (HR, 0.77), in a dose-response manner, Marilyn L. Kwan, PhD, and colleagues found.

Dr. Kwan is a senior research scientist at Kaiser Permanente Northern California Division of Research, Oakland.

Women with BMI less than 30 kg/m2 did not have a higher risk of mortality but a nonsignificant increase in the risk of recurrence was observed for those who consumed alcohol occasionally (HR, 1.29) and regularly (HR, 1.19), the investigators reported.

The findings were published online in Cancer.

Women included in the current study were participants in the Pathways Study and were diagnosed with stage I-IV breast cancer between 2003 and 2015. During follow-up, 524 recurrences and 834 deaths occurred, including 369 breast cancer-specific deaths, 314 cardiovascular disease-specific deaths, and 151 deaths from other health problems.

Alcohol consumption was assessed for the 6 months prior to cohort entry, which occurred at an average of about 2 months after diagnosis, as well as 6 months later – at an average of about 8 months after diagnosis – using a food-frequency questionnaire.

Compared with nondrinkers (36.9%), drinkers were more likely younger, more educated, and current or past smokers, the investigators noted.

“This profile appears counterintuitive yet might reflect a healthier lifestyle contributing to better overall survival. Furthermore, higher levels of alcohol consumption could lead to improvement in insulin sensitivity and reduction in insulin-like growth factor-1,” they speculated, noting that reduced fasting insulin concentrations and lower insulin-like growth factor-1 levels are linked with a decreased risk of type 2 diabetes, cardiovascular disease, and cancer.

“Many women with a history of breast cancer are interested in how to improve their prognosis and survival by making lifestyle changes after diagnosis,” they wrote, explaining the rationale for the study. “Current cancer prevention guidelines recommend avoiding alcohol intake or limiting consumption to no more than one drink per day for women. However, no specific guideline exists for cancer survivors other than following the cancer prevention guidelines to reduce the risk of a second cancer.”

High-quality studies on the impact of alcohol consumption on breast cancer prognosis are lacking, they added.

“Given that consuming alcohol is a potentially modifiable lifestyle factor after breast cancer diagnosis, further confirmation is warranted in other large prospective studies of breast cancer survivors with detailed exposure assessment and focus on body size,” they concluded.

The group is the first to report this finding in obese women, and they “strongly believe more research is needed to see if the same association is seen in other studies,” Dr. Kwan told this news organization.

“After a cancer diagnosis, many patients are motivated to make lifestyle changes,” she said. “That often includes adding exercise to their daily routine and eating a healthier diet. Our study findings suggest that doctors can tell patients that having up to a glass of alcohol a day is not likely to increase their risk of a breast cancer recurrence.”

This study was funded by the National Cancer Institute. The authors reported having no disclosures.
 

Drinking alcohol around the time of a breast cancer diagnosis may not have effects on prognosis that is mediated by body mass index (BMI), an analysis of data from a prospective cohort study suggests.

Kaiser Permanente
Dr. Marilyn Kwan

The study appears to show that drinking up to one serving of alcohol daily, including wine, beer, and liquor, was not associated with any specific outcomes after breast cancer diagnosis. The authors say these findings could have implications for developing more specific guidelines on alcohol use as it relates to the prevention of death and recurrence for cancer survivors.

Among 3,659 women followed for a mean of 11.2 years after a breast cancer diagnosis, overall alcohol consumption in the months before and up to 6 months after diagnosis was not associated with recurrence or mortality after adjusting for numerous factors such as age at diagnosis, cancer stage, socioeconomic details, smoking history, and preexisting conditions.

However, women with obesity (body mass index of 30 kg/m2 or greater) had a lower risk of mortality with increasing alcohol consumption for occasional drinking of 2 or more alcohol servings per week (hazard ratio, 0.71), and regular drinking of at least one alcohol serving daily (HR, 0.77), in a dose-response manner, Marilyn L. Kwan, PhD, and colleagues found.

Dr. Kwan is a senior research scientist at Kaiser Permanente Northern California Division of Research, Oakland.

Women with BMI less than 30 kg/m2 did not have a higher risk of mortality but a nonsignificant increase in the risk of recurrence was observed for those who consumed alcohol occasionally (HR, 1.29) and regularly (HR, 1.19), the investigators reported.

The findings were published online in Cancer.

Women included in the current study were participants in the Pathways Study and were diagnosed with stage I-IV breast cancer between 2003 and 2015. During follow-up, 524 recurrences and 834 deaths occurred, including 369 breast cancer-specific deaths, 314 cardiovascular disease-specific deaths, and 151 deaths from other health problems.

Alcohol consumption was assessed for the 6 months prior to cohort entry, which occurred at an average of about 2 months after diagnosis, as well as 6 months later – at an average of about 8 months after diagnosis – using a food-frequency questionnaire.

Compared with nondrinkers (36.9%), drinkers were more likely younger, more educated, and current or past smokers, the investigators noted.

“This profile appears counterintuitive yet might reflect a healthier lifestyle contributing to better overall survival. Furthermore, higher levels of alcohol consumption could lead to improvement in insulin sensitivity and reduction in insulin-like growth factor-1,” they speculated, noting that reduced fasting insulin concentrations and lower insulin-like growth factor-1 levels are linked with a decreased risk of type 2 diabetes, cardiovascular disease, and cancer.

“Many women with a history of breast cancer are interested in how to improve their prognosis and survival by making lifestyle changes after diagnosis,” they wrote, explaining the rationale for the study. “Current cancer prevention guidelines recommend avoiding alcohol intake or limiting consumption to no more than one drink per day for women. However, no specific guideline exists for cancer survivors other than following the cancer prevention guidelines to reduce the risk of a second cancer.”

High-quality studies on the impact of alcohol consumption on breast cancer prognosis are lacking, they added.

“Given that consuming alcohol is a potentially modifiable lifestyle factor after breast cancer diagnosis, further confirmation is warranted in other large prospective studies of breast cancer survivors with detailed exposure assessment and focus on body size,” they concluded.

The group is the first to report this finding in obese women, and they “strongly believe more research is needed to see if the same association is seen in other studies,” Dr. Kwan told this news organization.

“After a cancer diagnosis, many patients are motivated to make lifestyle changes,” she said. “That often includes adding exercise to their daily routine and eating a healthier diet. Our study findings suggest that doctors can tell patients that having up to a glass of alcohol a day is not likely to increase their risk of a breast cancer recurrence.”

This study was funded by the National Cancer Institute. The authors reported having no disclosures.
 

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM CANCER

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

FDA approves elranatamab for multiple myeloma

Article Type
Changed
Fri, 09/01/2023 - 17:14

The Food and Drug Administration has granted accelerated approval to the off-the-shelf biologic agent elranatamab (Elrexfio) for the treatment of relapsed or refractory multiple myeloma.

The B-cell maturation antigen (BCMA) CD3-targeted bispecific antibody (BsAb) was given Priority Review in February and had previously received Breakthrough Therapy Designation for relapsed or refractory multiple myeloma (RRMM), according to Pfizer.

Olivier Le Moal/Getty Images

FDA approval was based on favorable response and duration of response rates in the single-arm, phase 2 MagnetisMM-3 trial. The trial showed meaningful responses in heavily pretreated patients with RRMM who received elranatamab as their first BCMA-directed therapy.

The overall response rate in 97 BCMA-naive patients (cohort A) who previously received at least four lines of therapy, including a proteasome inhibitor, an immunomodulatory agent, and an anti-CD38 monoclonal antibody, was 58%, with an estimated 82% maintaining the response for 9 months or longer. Median time to first response was 1.2 months.

In 63 patients who received at least four prior lines of therapy, which also included a BCMA-directed therapy, the overall response rate was 33% after median follow-up of 10.2 months. An estimated 84% maintained a response for at least 9 months.

Elranatamab was given subcutaneously at a dose of 76 mg weekly on a 28-day cycle with a step-up priming dose regimen. The priming regimen included 12 mg and 32 mg doses on days 1 and 4, respectively, during cycle 1. Patients who received at least six cycles and showed at least a partial response for 2 or more months had a biweekly dosing interval.

Elranatamab carries a boxed warning for cytokine release syndrome (CRS) and neurologic toxicity, as well as warnings and precautions for infections, neutropenia, hepatotoxicity, and embryo–fetal toxicity. Therefore, the agent is available only through a restricted Risk Evaluation and Mitigation Strategy (REMS).

The boxed warning is included in the full prescribing information.

A confirmatory trial to gather additional safety and efficacy data was launched in 2022. Continued FDA approval is contingent on confirmed safety and efficacy data.

A version of this article first appeared on Medscape.com.

Publications
Topics
Sections

The Food and Drug Administration has granted accelerated approval to the off-the-shelf biologic agent elranatamab (Elrexfio) for the treatment of relapsed or refractory multiple myeloma.

The B-cell maturation antigen (BCMA) CD3-targeted bispecific antibody (BsAb) was given Priority Review in February and had previously received Breakthrough Therapy Designation for relapsed or refractory multiple myeloma (RRMM), according to Pfizer.

Olivier Le Moal/Getty Images

FDA approval was based on favorable response and duration of response rates in the single-arm, phase 2 MagnetisMM-3 trial. The trial showed meaningful responses in heavily pretreated patients with RRMM who received elranatamab as their first BCMA-directed therapy.

The overall response rate in 97 BCMA-naive patients (cohort A) who previously received at least four lines of therapy, including a proteasome inhibitor, an immunomodulatory agent, and an anti-CD38 monoclonal antibody, was 58%, with an estimated 82% maintaining the response for 9 months or longer. Median time to first response was 1.2 months.

In 63 patients who received at least four prior lines of therapy, which also included a BCMA-directed therapy, the overall response rate was 33% after median follow-up of 10.2 months. An estimated 84% maintained a response for at least 9 months.

Elranatamab was given subcutaneously at a dose of 76 mg weekly on a 28-day cycle with a step-up priming dose regimen. The priming regimen included 12 mg and 32 mg doses on days 1 and 4, respectively, during cycle 1. Patients who received at least six cycles and showed at least a partial response for 2 or more months had a biweekly dosing interval.

Elranatamab carries a boxed warning for cytokine release syndrome (CRS) and neurologic toxicity, as well as warnings and precautions for infections, neutropenia, hepatotoxicity, and embryo–fetal toxicity. Therefore, the agent is available only through a restricted Risk Evaluation and Mitigation Strategy (REMS).

The boxed warning is included in the full prescribing information.

A confirmatory trial to gather additional safety and efficacy data was launched in 2022. Continued FDA approval is contingent on confirmed safety and efficacy data.

A version of this article first appeared on Medscape.com.

The Food and Drug Administration has granted accelerated approval to the off-the-shelf biologic agent elranatamab (Elrexfio) for the treatment of relapsed or refractory multiple myeloma.

The B-cell maturation antigen (BCMA) CD3-targeted bispecific antibody (BsAb) was given Priority Review in February and had previously received Breakthrough Therapy Designation for relapsed or refractory multiple myeloma (RRMM), according to Pfizer.

Olivier Le Moal/Getty Images

FDA approval was based on favorable response and duration of response rates in the single-arm, phase 2 MagnetisMM-3 trial. The trial showed meaningful responses in heavily pretreated patients with RRMM who received elranatamab as their first BCMA-directed therapy.

The overall response rate in 97 BCMA-naive patients (cohort A) who previously received at least four lines of therapy, including a proteasome inhibitor, an immunomodulatory agent, and an anti-CD38 monoclonal antibody, was 58%, with an estimated 82% maintaining the response for 9 months or longer. Median time to first response was 1.2 months.

In 63 patients who received at least four prior lines of therapy, which also included a BCMA-directed therapy, the overall response rate was 33% after median follow-up of 10.2 months. An estimated 84% maintained a response for at least 9 months.

Elranatamab was given subcutaneously at a dose of 76 mg weekly on a 28-day cycle with a step-up priming dose regimen. The priming regimen included 12 mg and 32 mg doses on days 1 and 4, respectively, during cycle 1. Patients who received at least six cycles and showed at least a partial response for 2 or more months had a biweekly dosing interval.

Elranatamab carries a boxed warning for cytokine release syndrome (CRS) and neurologic toxicity, as well as warnings and precautions for infections, neutropenia, hepatotoxicity, and embryo–fetal toxicity. Therefore, the agent is available only through a restricted Risk Evaluation and Mitigation Strategy (REMS).

The boxed warning is included in the full prescribing information.

A confirmatory trial to gather additional safety and efficacy data was launched in 2022. Continued FDA approval is contingent on confirmed safety and efficacy data.

A version of this article first appeared on Medscape.com.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

FDA OKs combo therapy of niraparib, abiraterone acetate for prostate cancer

Article Type
Changed
Tue, 08/22/2023 - 11:27

The Food and Drug Administration has approved niraparib and abiraterone acetate (Akeega, Janssen Pharmaceuticals) to treat BRCA-positive, metastatic castration-resistant prostate cancer in adult patients with deleterious or suspected deleterious disease, as determined by an FDA-approved test.

The once-daily dual-action tablet is the first-and-only orally administered treatment combining the PARP inhibitor niraparib with abiraterone acetate.

Olivier Le Moal/Getty Images

The FDA’s approval was based on findings from the phase 3 MAGNITUDE precision medicine study, a randomized, placebo-controlled trial with 423 patients, 225 (53%) of whom had BRCA gene mutations as determined using a tissue assay such as FoundationOne CDx.

Among the subgroup with a BRCA mutation, radiographic progression-free survival was a median of 16.6 months vs. 10.9 months (hazard ratio [HR], 0.53; 95% confidence interval [CI], 0.36-0.79; P = .0014). In this subgroup, an exploratory overall survival analysis demonstrated a median of 30.4 months vs. 28.6 months (HR, 0.79; 95% CI, 0.55-1.12), favoring the treatment arm.

Although the overall cohort (those with and without BRCA mutations) demonstrated a significant improvement in radiographic progression-free survival, the subgroup with non-BRCA homologous recombination repair mutations did not demonstrate a significant improvement in radiographic progression-free survival, which indicates that the benefit observed was “primarily attributed” to the results in the subgroup of patients with BRCA mutations, according to the FDA.

The safety profile of niraparib and abiraterone acetate plus prednisone was consistent with the known safety profile of each FDA-approved monotherapy. Serious adverse events occurred in 41% of patients in the treatment arm. These most often included musculoskeletal pain (44% vs. 42%), fatigue (43% vs. 30%), constipation (34% vs. 20%), hypertension (33% vs. 27%), and nausea (33% vs. 21%).

An adverse reaction led to permanent discontinuation of treatment in 15% of patients.

“As a physician, identifying patients with a worse prognosis is a priority, especially those whose cancers have a BRCA mutation,” principal investigator Kim Chi, MD, stated in the Janssen press release. “We prospectively designed the MAGNITUDE study to identify the subset of patients most likely to benefit from targeted treatment with AKEEGA and to help us understand how we can potentially achieve better health outcomes for patients.”

About 10%-15% of patients who develop metastatic castration-resistant prostate cancer have BRCA gene alterations, and those patients are more likely to have aggressive disease, poor outcomes, and shorter survival. Therefore, this new agent “brings an important treatment option to patients with prostate cancer as they consider their road ahead,” said Shelby Moneer, vice president of patient programs and education at ZERO Prostate Cancer.

The prescribing information lists the recommended dose at 200 mg niraparib and 1,000 mg abiraterone once daily in combination with 10 mg of prednisone daily until disease progression or unacceptable toxicity. Patients should also receive a gonadotropin-releasing hormone analog concurrently or should have had bilateral orchiectomy.

Health care professionals should report all serious adverse events suspected to be associated with the use of any medicine and device by using the FDA’s MedWatch Reporting System or by calling 1-800-FDA-1088.

A version of this article appeared on Medscape.com.

Publications
Topics
Sections

The Food and Drug Administration has approved niraparib and abiraterone acetate (Akeega, Janssen Pharmaceuticals) to treat BRCA-positive, metastatic castration-resistant prostate cancer in adult patients with deleterious or suspected deleterious disease, as determined by an FDA-approved test.

The once-daily dual-action tablet is the first-and-only orally administered treatment combining the PARP inhibitor niraparib with abiraterone acetate.

Olivier Le Moal/Getty Images

The FDA’s approval was based on findings from the phase 3 MAGNITUDE precision medicine study, a randomized, placebo-controlled trial with 423 patients, 225 (53%) of whom had BRCA gene mutations as determined using a tissue assay such as FoundationOne CDx.

Among the subgroup with a BRCA mutation, radiographic progression-free survival was a median of 16.6 months vs. 10.9 months (hazard ratio [HR], 0.53; 95% confidence interval [CI], 0.36-0.79; P = .0014). In this subgroup, an exploratory overall survival analysis demonstrated a median of 30.4 months vs. 28.6 months (HR, 0.79; 95% CI, 0.55-1.12), favoring the treatment arm.

Although the overall cohort (those with and without BRCA mutations) demonstrated a significant improvement in radiographic progression-free survival, the subgroup with non-BRCA homologous recombination repair mutations did not demonstrate a significant improvement in radiographic progression-free survival, which indicates that the benefit observed was “primarily attributed” to the results in the subgroup of patients with BRCA mutations, according to the FDA.

The safety profile of niraparib and abiraterone acetate plus prednisone was consistent with the known safety profile of each FDA-approved monotherapy. Serious adverse events occurred in 41% of patients in the treatment arm. These most often included musculoskeletal pain (44% vs. 42%), fatigue (43% vs. 30%), constipation (34% vs. 20%), hypertension (33% vs. 27%), and nausea (33% vs. 21%).

An adverse reaction led to permanent discontinuation of treatment in 15% of patients.

“As a physician, identifying patients with a worse prognosis is a priority, especially those whose cancers have a BRCA mutation,” principal investigator Kim Chi, MD, stated in the Janssen press release. “We prospectively designed the MAGNITUDE study to identify the subset of patients most likely to benefit from targeted treatment with AKEEGA and to help us understand how we can potentially achieve better health outcomes for patients.”

About 10%-15% of patients who develop metastatic castration-resistant prostate cancer have BRCA gene alterations, and those patients are more likely to have aggressive disease, poor outcomes, and shorter survival. Therefore, this new agent “brings an important treatment option to patients with prostate cancer as they consider their road ahead,” said Shelby Moneer, vice president of patient programs and education at ZERO Prostate Cancer.

The prescribing information lists the recommended dose at 200 mg niraparib and 1,000 mg abiraterone once daily in combination with 10 mg of prednisone daily until disease progression or unacceptable toxicity. Patients should also receive a gonadotropin-releasing hormone analog concurrently or should have had bilateral orchiectomy.

Health care professionals should report all serious adverse events suspected to be associated with the use of any medicine and device by using the FDA’s MedWatch Reporting System or by calling 1-800-FDA-1088.

A version of this article appeared on Medscape.com.

The Food and Drug Administration has approved niraparib and abiraterone acetate (Akeega, Janssen Pharmaceuticals) to treat BRCA-positive, metastatic castration-resistant prostate cancer in adult patients with deleterious or suspected deleterious disease, as determined by an FDA-approved test.

The once-daily dual-action tablet is the first-and-only orally administered treatment combining the PARP inhibitor niraparib with abiraterone acetate.

Olivier Le Moal/Getty Images

The FDA’s approval was based on findings from the phase 3 MAGNITUDE precision medicine study, a randomized, placebo-controlled trial with 423 patients, 225 (53%) of whom had BRCA gene mutations as determined using a tissue assay such as FoundationOne CDx.

Among the subgroup with a BRCA mutation, radiographic progression-free survival was a median of 16.6 months vs. 10.9 months (hazard ratio [HR], 0.53; 95% confidence interval [CI], 0.36-0.79; P = .0014). In this subgroup, an exploratory overall survival analysis demonstrated a median of 30.4 months vs. 28.6 months (HR, 0.79; 95% CI, 0.55-1.12), favoring the treatment arm.

Although the overall cohort (those with and without BRCA mutations) demonstrated a significant improvement in radiographic progression-free survival, the subgroup with non-BRCA homologous recombination repair mutations did not demonstrate a significant improvement in radiographic progression-free survival, which indicates that the benefit observed was “primarily attributed” to the results in the subgroup of patients with BRCA mutations, according to the FDA.

The safety profile of niraparib and abiraterone acetate plus prednisone was consistent with the known safety profile of each FDA-approved monotherapy. Serious adverse events occurred in 41% of patients in the treatment arm. These most often included musculoskeletal pain (44% vs. 42%), fatigue (43% vs. 30%), constipation (34% vs. 20%), hypertension (33% vs. 27%), and nausea (33% vs. 21%).

An adverse reaction led to permanent discontinuation of treatment in 15% of patients.

“As a physician, identifying patients with a worse prognosis is a priority, especially those whose cancers have a BRCA mutation,” principal investigator Kim Chi, MD, stated in the Janssen press release. “We prospectively designed the MAGNITUDE study to identify the subset of patients most likely to benefit from targeted treatment with AKEEGA and to help us understand how we can potentially achieve better health outcomes for patients.”

About 10%-15% of patients who develop metastatic castration-resistant prostate cancer have BRCA gene alterations, and those patients are more likely to have aggressive disease, poor outcomes, and shorter survival. Therefore, this new agent “brings an important treatment option to patients with prostate cancer as they consider their road ahead,” said Shelby Moneer, vice president of patient programs and education at ZERO Prostate Cancer.

The prescribing information lists the recommended dose at 200 mg niraparib and 1,000 mg abiraterone once daily in combination with 10 mg of prednisone daily until disease progression or unacceptable toxicity. Patients should also receive a gonadotropin-releasing hormone analog concurrently or should have had bilateral orchiectomy.

Health care professionals should report all serious adverse events suspected to be associated with the use of any medicine and device by using the FDA’s MedWatch Reporting System or by calling 1-800-FDA-1088.

A version of this article appeared on Medscape.com.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

FDA OKs talquetamab, a first-in-class myeloma tx

Article Type
Changed
Wed, 08/16/2023 - 17:17

 

The U.S. Food and Drug Administration (FDA) has granted accelerated approval to talquetamab-tgvs (Talvey, Janssen Biotech, Inc), a first-in-class bispecific antibody targeting the GPRC5D receptor, for heavily pretreated adults with relapsed or refractory multiple myeloma.

Patients must have received at least four prior lines of therapy, including a proteasome inhibitor, an immunomodulatory agent, and an anti-CD38 monoclonal antibody.

The agent, which also received breakthrough and orphan drug designation, is available only through the Tecvayli-Talvey Risk Evaluation and Mitigation Strategy (REMS) because of a boxed warning for life-threatening or fatal cytokine release syndrome (CRS) and neurological toxicity, including immune effector cell–associated neurotoxicity (ICANS), the FDA announced.

Talquetamab-tgvs was evaluated in the single-arm, open-label MonumenTAL-1 study of 187 patients who had previously been treated with at least four prior systemic therapies.

The overall response rate in 100 patients who received a subcutaneous dose of 0.4 mg/kg weekly was 73% and median duration of response was 9.5 months. The overall response rate in 87 patients who received a subcutaneous dose of 0.8 mg/kg biweekly was 73.6%, with about 85% of responders maintaining their response for at least 9 months. In this group, the median duration of response was not estimable.

Patients in the 0.4 mg/kg weekly dose group were treated following two step-up doses in the first week of therapy, and those in the 0.8 mg/kg biweekly group were treated following three step-up doses, until disease progression or unacceptable toxicity.

Adverse reactions occurring in at least 20% of the 339 patients in the safety population included CRS, dysgeusia (foul, metallic taste sensation), nail disorder, musculoskeletal pain, skin disorder, rash, fatigue, decreased weight, dry mouth, pyrexia, xerosis, dysphagia, upper respiratory tract infection, and diarrhea.

Both the weekly 0.4 mg/kg and biweekly 0.8 mg/kg doses are recommended. The full dosing schedule is included in the prescribing information.

The approval follows a series of market withdrawals for other multiple myeloma drugs that initially received accelerated FDA approval. For instance, the FDA recently requested withdrawal of melphalan flufenamide (Pepaxto) after 2021 confirmatory trial results showed an increased risk of death. This agent had received accelerated approval in 2021. GlaxoSmithKline’s blood cancer drugs panobinostat (Farydak) and belantamab mafodotin-blmf (Blenrep) were also withdrawn based on confirmatory trial results.

Continued approval of talquetemab-tgvs for this indication is also contingent on verifying efficacy in confirmatory trials.

The new treatment approach represents a “welcome addition to the myeloma community,” Michael Andreini, president and chief executive officer of the Multiple Myeloma Research Foundation stated in a Janssen press release. “Although options for the treatment of multiple myeloma have expanded significantly in recent years, the disease remains incurable, and therefore, patients are in need of new treatment options.”

Health care professionals should report all serious adverse events suspected to be associated with the use of any medicine and device to FDA’s MedWatch Reporting System or by calling 1-800-FDA-1088.

A version of this article first appeared on Medscape.com.

Publications
Topics
Sections

 

The U.S. Food and Drug Administration (FDA) has granted accelerated approval to talquetamab-tgvs (Talvey, Janssen Biotech, Inc), a first-in-class bispecific antibody targeting the GPRC5D receptor, for heavily pretreated adults with relapsed or refractory multiple myeloma.

Patients must have received at least four prior lines of therapy, including a proteasome inhibitor, an immunomodulatory agent, and an anti-CD38 monoclonal antibody.

The agent, which also received breakthrough and orphan drug designation, is available only through the Tecvayli-Talvey Risk Evaluation and Mitigation Strategy (REMS) because of a boxed warning for life-threatening or fatal cytokine release syndrome (CRS) and neurological toxicity, including immune effector cell–associated neurotoxicity (ICANS), the FDA announced.

Talquetamab-tgvs was evaluated in the single-arm, open-label MonumenTAL-1 study of 187 patients who had previously been treated with at least four prior systemic therapies.

The overall response rate in 100 patients who received a subcutaneous dose of 0.4 mg/kg weekly was 73% and median duration of response was 9.5 months. The overall response rate in 87 patients who received a subcutaneous dose of 0.8 mg/kg biweekly was 73.6%, with about 85% of responders maintaining their response for at least 9 months. In this group, the median duration of response was not estimable.

Patients in the 0.4 mg/kg weekly dose group were treated following two step-up doses in the first week of therapy, and those in the 0.8 mg/kg biweekly group were treated following three step-up doses, until disease progression or unacceptable toxicity.

Adverse reactions occurring in at least 20% of the 339 patients in the safety population included CRS, dysgeusia (foul, metallic taste sensation), nail disorder, musculoskeletal pain, skin disorder, rash, fatigue, decreased weight, dry mouth, pyrexia, xerosis, dysphagia, upper respiratory tract infection, and diarrhea.

Both the weekly 0.4 mg/kg and biweekly 0.8 mg/kg doses are recommended. The full dosing schedule is included in the prescribing information.

The approval follows a series of market withdrawals for other multiple myeloma drugs that initially received accelerated FDA approval. For instance, the FDA recently requested withdrawal of melphalan flufenamide (Pepaxto) after 2021 confirmatory trial results showed an increased risk of death. This agent had received accelerated approval in 2021. GlaxoSmithKline’s blood cancer drugs panobinostat (Farydak) and belantamab mafodotin-blmf (Blenrep) were also withdrawn based on confirmatory trial results.

Continued approval of talquetemab-tgvs for this indication is also contingent on verifying efficacy in confirmatory trials.

The new treatment approach represents a “welcome addition to the myeloma community,” Michael Andreini, president and chief executive officer of the Multiple Myeloma Research Foundation stated in a Janssen press release. “Although options for the treatment of multiple myeloma have expanded significantly in recent years, the disease remains incurable, and therefore, patients are in need of new treatment options.”

Health care professionals should report all serious adverse events suspected to be associated with the use of any medicine and device to FDA’s MedWatch Reporting System or by calling 1-800-FDA-1088.

A version of this article first appeared on Medscape.com.

 

The U.S. Food and Drug Administration (FDA) has granted accelerated approval to talquetamab-tgvs (Talvey, Janssen Biotech, Inc), a first-in-class bispecific antibody targeting the GPRC5D receptor, for heavily pretreated adults with relapsed or refractory multiple myeloma.

Patients must have received at least four prior lines of therapy, including a proteasome inhibitor, an immunomodulatory agent, and an anti-CD38 monoclonal antibody.

The agent, which also received breakthrough and orphan drug designation, is available only through the Tecvayli-Talvey Risk Evaluation and Mitigation Strategy (REMS) because of a boxed warning for life-threatening or fatal cytokine release syndrome (CRS) and neurological toxicity, including immune effector cell–associated neurotoxicity (ICANS), the FDA announced.

Talquetamab-tgvs was evaluated in the single-arm, open-label MonumenTAL-1 study of 187 patients who had previously been treated with at least four prior systemic therapies.

The overall response rate in 100 patients who received a subcutaneous dose of 0.4 mg/kg weekly was 73% and median duration of response was 9.5 months. The overall response rate in 87 patients who received a subcutaneous dose of 0.8 mg/kg biweekly was 73.6%, with about 85% of responders maintaining their response for at least 9 months. In this group, the median duration of response was not estimable.

Patients in the 0.4 mg/kg weekly dose group were treated following two step-up doses in the first week of therapy, and those in the 0.8 mg/kg biweekly group were treated following three step-up doses, until disease progression or unacceptable toxicity.

Adverse reactions occurring in at least 20% of the 339 patients in the safety population included CRS, dysgeusia (foul, metallic taste sensation), nail disorder, musculoskeletal pain, skin disorder, rash, fatigue, decreased weight, dry mouth, pyrexia, xerosis, dysphagia, upper respiratory tract infection, and diarrhea.

Both the weekly 0.4 mg/kg and biweekly 0.8 mg/kg doses are recommended. The full dosing schedule is included in the prescribing information.

The approval follows a series of market withdrawals for other multiple myeloma drugs that initially received accelerated FDA approval. For instance, the FDA recently requested withdrawal of melphalan flufenamide (Pepaxto) after 2021 confirmatory trial results showed an increased risk of death. This agent had received accelerated approval in 2021. GlaxoSmithKline’s blood cancer drugs panobinostat (Farydak) and belantamab mafodotin-blmf (Blenrep) were also withdrawn based on confirmatory trial results.

Continued approval of talquetemab-tgvs for this indication is also contingent on verifying efficacy in confirmatory trials.

The new treatment approach represents a “welcome addition to the myeloma community,” Michael Andreini, president and chief executive officer of the Multiple Myeloma Research Foundation stated in a Janssen press release. “Although options for the treatment of multiple myeloma have expanded significantly in recent years, the disease remains incurable, and therefore, patients are in need of new treatment options.”

Health care professionals should report all serious adverse events suspected to be associated with the use of any medicine and device to FDA’s MedWatch Reporting System or by calling 1-800-FDA-1088.

A version of this article first appeared on Medscape.com.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

AI-supported breast screens may reduce radiologist workload

Article Type
Changed
Fri, 08/04/2023 - 09:52

Artificial intelligence (AI)–supported breast cancer screening appears safe and at least as accurate as standard double reading of mammograms by two breast radiologists, according to early results from a large, randomized, population-based cohort study.

The AI-supported screening also reduced radiologist workload by nearly 44%, researchers estimated.

The trial also found a 20% increase in cancer detection using AI support compared with routine double mammography reading, underscoring AI’s potential to improve screening accuracy and efficiency.

The findings, published online in Lancet Oncology, come from a planned interim safety analysis of the Swedish Mammography Screening with Artificial Intelligence (MASAI) trial.

To date, AI has shown promise in mammography screening, with retrospective evidence demonstrating similar accuracy, compared with standard double readings as well as reduced workload for radiologists. Still, randomized trials assessing the efficacy of AI-supported breast screening are needed.

The aim of the current interim randomized analysis was to assess early screening performance, which included cancer detection, recall, and false positive rates as well as cancer type detected and workload.

The MASAI trial randomized 80,033 women, with a median age of 54, to AI-supported screening (n = 40,003) or double reading without AI (n = 40,030).

The AI system provided malignancy risk scores from 1 to 10, with low-risk scores ranging from 1 to 7, intermediate risk from 8 to 9, and high risk at 10. These risk scores were used to triage screening exams to a single radiologist reading (score of 1-9) or double reading (score of 10), given that cancer prevalence “increases sharply” for those with a risk score of 10, the researchers explained. The AI system also provided computer-aided detection marks for exams with risk scores of 8-10 to radiologists.

Among nearly 40,000 women screened with AI support, 244 cancers were detected, including 184 invasive cancers (75%) and 60 in situ cancers (25%), and resulted in 861 recalls. Among 40,024 participants receiving standard screening, radiologists detected 203 cancers, including 165 invasive cancers (81%) and 38 in situ cancers (19%), and resulted in 817 recalls.

Overall, the detection rate using AI support versus standard screening was 6.1 per 1000 screened participants versus 5.1 per 1,000. The recall rates were 2.2% versus 2.0%, respectively.

The false positive rates were the same in both groups (1.5%) while the positive predictive value (PPV) of recall – how likely a recall of a participant ultimately led to a cancer diagnosis – was higher in the AI group: 28.3% versus 24.8%.

The cancer detection rate in the high-risk group – patients with a risk score of 10 – was 72.3 per 1000 participants screened, or one cancer per 14 screening exams. And, overall, 189 of 490 screening exams flagged as extra-high risk by AI (the highest 1% risk) were recalled. Of the 189 recalled participants, 136 had cancer, representing a PPV of recall of 72%.

Overall, “we found that the benefit of AI-supported screening in terms of screen-reading workload reduction was considerable,” the authors said.

Assuming a radiologist can read 50 mammograms an hour, the researchers estimated that a radiologist would take 4.6 fewer months to read more than 46,000 screening exams in the intervention group compared with more than 83,000 in the control group.

Although these early safety results are “promising,” the findings “are not enough on their own to confirm that AI is ready to be implemented in mammography screening,” lead author Kristina Lång, PhD, of Lund (Sweden) University, said in a press release.

“We still need to understand the implications on patients’ outcomes, especially whether combining radiologists’ expertise with AI can help detect interval cancers that are often missed by traditional screening, as well as the cost-effectiveness of the technology,” she said, adding that “the greatest potential of AI right now is that it could allow radiologists to be less burdened by the excessive amount of reading.”

In an accompanying editorial, Nereo Segnan, MD, and Antonio Ponti, MD, both of CPO Piemonte in Torino, Italy, said that the AI risk score for breast cancer in the trial “seems very accurate at being able to separate high-risk from low-risk women.”

However, the potential for overdiagnosis or overdetection of indolent lesions in the intervention group should “prompt caution in the interpretation of results that otherwise seem straightforward in favoring the use of AI,” the editorialists noted.

The authors agreed that increased detection of in situ cancers with AI-supported screening compared with standard screening – 25% versus 19% – “could be concerning in terms of overdiagnosis,” as the risk of overtreatment is more likely with these low-grade cancers.

In the final analysis, Dr. Lång and colleagues plan to characterize the biological features of detected lesions to provide further insight on AI-supported screening, including the risk for overdiagnosis.

In a statement to the U.K.-based Science Media Centre, Stephen Duffy, professor of cancer screening, Wolfson Institute of Population Health, Queen Mary University of London, commented that the “results illustrate the potential for artificial intelligence to reduce the burden on radiologists’ time,” which is “an issue of considerable importance in many breast screening programs.”

The MASAI study was funded by the Swedish Cancer Society, Confederation of Regional Cancer Centres, and government funding for clinical research. Dr. Lång has been an advisory board member for Siemens Healthineers and has received lecture honorarium from AstraZeneca. Dr. Segnan and Dr. Hall reported no relevant financial relationships.

A version of this article first appeared on Medscape.com.

Publications
Topics
Sections

Artificial intelligence (AI)–supported breast cancer screening appears safe and at least as accurate as standard double reading of mammograms by two breast radiologists, according to early results from a large, randomized, population-based cohort study.

The AI-supported screening also reduced radiologist workload by nearly 44%, researchers estimated.

The trial also found a 20% increase in cancer detection using AI support compared with routine double mammography reading, underscoring AI’s potential to improve screening accuracy and efficiency.

The findings, published online in Lancet Oncology, come from a planned interim safety analysis of the Swedish Mammography Screening with Artificial Intelligence (MASAI) trial.

To date, AI has shown promise in mammography screening, with retrospective evidence demonstrating similar accuracy, compared with standard double readings as well as reduced workload for radiologists. Still, randomized trials assessing the efficacy of AI-supported breast screening are needed.

The aim of the current interim randomized analysis was to assess early screening performance, which included cancer detection, recall, and false positive rates as well as cancer type detected and workload.

The MASAI trial randomized 80,033 women, with a median age of 54, to AI-supported screening (n = 40,003) or double reading without AI (n = 40,030).

The AI system provided malignancy risk scores from 1 to 10, with low-risk scores ranging from 1 to 7, intermediate risk from 8 to 9, and high risk at 10. These risk scores were used to triage screening exams to a single radiologist reading (score of 1-9) or double reading (score of 10), given that cancer prevalence “increases sharply” for those with a risk score of 10, the researchers explained. The AI system also provided computer-aided detection marks for exams with risk scores of 8-10 to radiologists.

Among nearly 40,000 women screened with AI support, 244 cancers were detected, including 184 invasive cancers (75%) and 60 in situ cancers (25%), and resulted in 861 recalls. Among 40,024 participants receiving standard screening, radiologists detected 203 cancers, including 165 invasive cancers (81%) and 38 in situ cancers (19%), and resulted in 817 recalls.

Overall, the detection rate using AI support versus standard screening was 6.1 per 1000 screened participants versus 5.1 per 1,000. The recall rates were 2.2% versus 2.0%, respectively.

The false positive rates were the same in both groups (1.5%) while the positive predictive value (PPV) of recall – how likely a recall of a participant ultimately led to a cancer diagnosis – was higher in the AI group: 28.3% versus 24.8%.

The cancer detection rate in the high-risk group – patients with a risk score of 10 – was 72.3 per 1000 participants screened, or one cancer per 14 screening exams. And, overall, 189 of 490 screening exams flagged as extra-high risk by AI (the highest 1% risk) were recalled. Of the 189 recalled participants, 136 had cancer, representing a PPV of recall of 72%.

Overall, “we found that the benefit of AI-supported screening in terms of screen-reading workload reduction was considerable,” the authors said.

Assuming a radiologist can read 50 mammograms an hour, the researchers estimated that a radiologist would take 4.6 fewer months to read more than 46,000 screening exams in the intervention group compared with more than 83,000 in the control group.

Although these early safety results are “promising,” the findings “are not enough on their own to confirm that AI is ready to be implemented in mammography screening,” lead author Kristina Lång, PhD, of Lund (Sweden) University, said in a press release.

“We still need to understand the implications on patients’ outcomes, especially whether combining radiologists’ expertise with AI can help detect interval cancers that are often missed by traditional screening, as well as the cost-effectiveness of the technology,” she said, adding that “the greatest potential of AI right now is that it could allow radiologists to be less burdened by the excessive amount of reading.”

In an accompanying editorial, Nereo Segnan, MD, and Antonio Ponti, MD, both of CPO Piemonte in Torino, Italy, said that the AI risk score for breast cancer in the trial “seems very accurate at being able to separate high-risk from low-risk women.”

However, the potential for overdiagnosis or overdetection of indolent lesions in the intervention group should “prompt caution in the interpretation of results that otherwise seem straightforward in favoring the use of AI,” the editorialists noted.

The authors agreed that increased detection of in situ cancers with AI-supported screening compared with standard screening – 25% versus 19% – “could be concerning in terms of overdiagnosis,” as the risk of overtreatment is more likely with these low-grade cancers.

In the final analysis, Dr. Lång and colleagues plan to characterize the biological features of detected lesions to provide further insight on AI-supported screening, including the risk for overdiagnosis.

In a statement to the U.K.-based Science Media Centre, Stephen Duffy, professor of cancer screening, Wolfson Institute of Population Health, Queen Mary University of London, commented that the “results illustrate the potential for artificial intelligence to reduce the burden on radiologists’ time,” which is “an issue of considerable importance in many breast screening programs.”

The MASAI study was funded by the Swedish Cancer Society, Confederation of Regional Cancer Centres, and government funding for clinical research. Dr. Lång has been an advisory board member for Siemens Healthineers and has received lecture honorarium from AstraZeneca. Dr. Segnan and Dr. Hall reported no relevant financial relationships.

A version of this article first appeared on Medscape.com.

Artificial intelligence (AI)–supported breast cancer screening appears safe and at least as accurate as standard double reading of mammograms by two breast radiologists, according to early results from a large, randomized, population-based cohort study.

The AI-supported screening also reduced radiologist workload by nearly 44%, researchers estimated.

The trial also found a 20% increase in cancer detection using AI support compared with routine double mammography reading, underscoring AI’s potential to improve screening accuracy and efficiency.

The findings, published online in Lancet Oncology, come from a planned interim safety analysis of the Swedish Mammography Screening with Artificial Intelligence (MASAI) trial.

To date, AI has shown promise in mammography screening, with retrospective evidence demonstrating similar accuracy, compared with standard double readings as well as reduced workload for radiologists. Still, randomized trials assessing the efficacy of AI-supported breast screening are needed.

The aim of the current interim randomized analysis was to assess early screening performance, which included cancer detection, recall, and false positive rates as well as cancer type detected and workload.

The MASAI trial randomized 80,033 women, with a median age of 54, to AI-supported screening (n = 40,003) or double reading without AI (n = 40,030).

The AI system provided malignancy risk scores from 1 to 10, with low-risk scores ranging from 1 to 7, intermediate risk from 8 to 9, and high risk at 10. These risk scores were used to triage screening exams to a single radiologist reading (score of 1-9) or double reading (score of 10), given that cancer prevalence “increases sharply” for those with a risk score of 10, the researchers explained. The AI system also provided computer-aided detection marks for exams with risk scores of 8-10 to radiologists.

Among nearly 40,000 women screened with AI support, 244 cancers were detected, including 184 invasive cancers (75%) and 60 in situ cancers (25%), and resulted in 861 recalls. Among 40,024 participants receiving standard screening, radiologists detected 203 cancers, including 165 invasive cancers (81%) and 38 in situ cancers (19%), and resulted in 817 recalls.

Overall, the detection rate using AI support versus standard screening was 6.1 per 1000 screened participants versus 5.1 per 1,000. The recall rates were 2.2% versus 2.0%, respectively.

The false positive rates were the same in both groups (1.5%) while the positive predictive value (PPV) of recall – how likely a recall of a participant ultimately led to a cancer diagnosis – was higher in the AI group: 28.3% versus 24.8%.

The cancer detection rate in the high-risk group – patients with a risk score of 10 – was 72.3 per 1000 participants screened, or one cancer per 14 screening exams. And, overall, 189 of 490 screening exams flagged as extra-high risk by AI (the highest 1% risk) were recalled. Of the 189 recalled participants, 136 had cancer, representing a PPV of recall of 72%.

Overall, “we found that the benefit of AI-supported screening in terms of screen-reading workload reduction was considerable,” the authors said.

Assuming a radiologist can read 50 mammograms an hour, the researchers estimated that a radiologist would take 4.6 fewer months to read more than 46,000 screening exams in the intervention group compared with more than 83,000 in the control group.

Although these early safety results are “promising,” the findings “are not enough on their own to confirm that AI is ready to be implemented in mammography screening,” lead author Kristina Lång, PhD, of Lund (Sweden) University, said in a press release.

“We still need to understand the implications on patients’ outcomes, especially whether combining radiologists’ expertise with AI can help detect interval cancers that are often missed by traditional screening, as well as the cost-effectiveness of the technology,” she said, adding that “the greatest potential of AI right now is that it could allow radiologists to be less burdened by the excessive amount of reading.”

In an accompanying editorial, Nereo Segnan, MD, and Antonio Ponti, MD, both of CPO Piemonte in Torino, Italy, said that the AI risk score for breast cancer in the trial “seems very accurate at being able to separate high-risk from low-risk women.”

However, the potential for overdiagnosis or overdetection of indolent lesions in the intervention group should “prompt caution in the interpretation of results that otherwise seem straightforward in favoring the use of AI,” the editorialists noted.

The authors agreed that increased detection of in situ cancers with AI-supported screening compared with standard screening – 25% versus 19% – “could be concerning in terms of overdiagnosis,” as the risk of overtreatment is more likely with these low-grade cancers.

In the final analysis, Dr. Lång and colleagues plan to characterize the biological features of detected lesions to provide further insight on AI-supported screening, including the risk for overdiagnosis.

In a statement to the U.K.-based Science Media Centre, Stephen Duffy, professor of cancer screening, Wolfson Institute of Population Health, Queen Mary University of London, commented that the “results illustrate the potential for artificial intelligence to reduce the burden on radiologists’ time,” which is “an issue of considerable importance in many breast screening programs.”

The MASAI study was funded by the Swedish Cancer Society, Confederation of Regional Cancer Centres, and government funding for clinical research. Dr. Lång has been an advisory board member for Siemens Healthineers and has received lecture honorarium from AstraZeneca. Dr. Segnan and Dr. Hall reported no relevant financial relationships.

A version of this article first appeared on Medscape.com.

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM LANCET ONCOLOGY

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Oncologists challenge ‘burdensome’ MOC requirements

Article Type
Changed
Wed, 08/02/2023 - 12:40

A petition demanding an end to maintenance of certification (MOC) requirements from the American Board of Internal Medicine (ABIM) gained traction among oncologists in late July, garnering nearly 7,500 signatures in 10 days.

The MOC program, “originally intended to uphold the standards of medical practice and promote lifelong learning, has evolved into a complex and time-consuming process that poses significant challenges to practicing physicians,” according to the petition launched on July 21 by hematologist-oncologist Aaron Goodman, MD. The MOC “has become burdensome, costly, and lacks evidence to support its effectiveness in improving patient care or physician competence.”

Dr. Goodman, assistant professor at the University of California, San Diego, is scheduled to debate the matter with ABIM President and Chief Executive Officer Richard J. Baron, MD, during a Healthcare Unfiltered podcast recorded and hosted by Chadi Nabhan, MD. In the August 3 podcast, Dr. Goodman and Dr. Baron will respond to questions posed via tweets, Dr. Goodman said.

Twitter survey posted by Dr. Nabhan in advance of the debate asked physicians whether the cost of the MOC, the time required for testing, or data sharing by ABIM is most bothersome. Of 158 respondents, 71% selected “all of the above.”
 

ABIM touts MOC ‘values’  

The ABIM requires an initial certification assessment that costs thousands of dollars and must be repeated every 10 years. The annual MOC requirements, which were tacked on within the last decade, involve tests that cost $220 for the first certificate a physician holds and about $120 for each subsequent one.

Over the course of his career, Dr. Goodman estimates he will spend over $40,000 to maintain his three ABIM boards in medicine, hematology, and oncology.

The ABIM did not immediately respond to a request for comment on the petition and debate, but a page on the ABIM website touts the “values of MOC” and says there is “compelling evidence” that the MOC improves the value of care without sacrificing quality and that board-certified physicians earn more.

According to the website, the MOC program “provides doctors with a pathway to know that they are staying current in the medical knowledge they use to treat patients and make important care decisions daily.” The ABIM also says that the “program has evolved to include new assessment options and an increased recognition of the work doctors do every day” and that the ABIM “continuously collaborates with doctors to increase the relevance of exams.”

Aniruddha Singh, MD, also weighed in on the value of MOC in a July 13 ABIM blog post. Dr. Singh is a member of this year’s Cardiovascular Disease Traditional, 10-Year MOC Exam Approval Committee and program director for the General Cardiovascular Fellowship at Drexel University College of Medicine, Reading, Pa.

In his post, Dr. Singh states that the MOC “facilitates a broader perspective on a range of topics [and] enables me to delve deeper into relevant areas, fostering a comprehensive understanding that enhances my quality of care.”
 

Growing resistance

Although Dr. Goodman acknowledged the merit of board testing every decade or so, physicians already do continuing medical education (CME) to keep up-to-date on their specialties. Dr. Goodman believes that most physicians, other than those who work for ABIM, would agree that MOC is a waste of time and money.

“It’s a pain-in-the-ass module that you sit at home and Google – it’s not really any sort of assessment,” nor does it help protect the public, said Goodman. The MOC is ultimately “just a money grab.”

According to the ABIM’s website, the nonprofit has net assets of more than $73.2 million as of June 30, 2022. Last year, the ABIM’s revenue hit $71.9 million, with more than half coming from MOC fees and 48% from certification.

The ABIM also says it spent $58 million in 2022. A breakdown shows about 63% of that money went to administering (28%), researching (13%), overseeing (4%), and developing (18%) the certification and MOC exams and program. ABIM’s CEO Dr. Baron makes about $1.2 million a year, according to recent tax filings. The COO makes about $550,000 from the ABIM and “related” organizations.

Fed up with the requirements and cost, Dr. Goodman decided to launch the petition to see if others agreed and how many.

His petition, addressed to the ABIM, expresses “deep dissatisfaction” with the ABIM MOC program and “respectfully request[s] that the ABIM take immediate action to eliminate the MOC program and adopt alternative, less burdensome methods of ensuring physician competence and continuous professional development.” Dr. Goodman, alongside Vinay Prasad, MD, a hematologist-oncologist and professor at the University of California, San Francisco, reiterated these points in a piece highlighting the petition.

Shortly before the petition went public but after he had been vocal on Twitter about issues with the MOC, Dr. Goodman said he received an invitation to join the ABIM Board of Governors. “My hypothesis is they are trying to ‘friend’ me, so I get credentialed, and they get me to stop yelling. It just made me more pissed off. I don’t want to be any part of that,” he said.

H. Jack West, MD, a thoracic oncologist and associate professor at City of Hope Comprehensive Cancer Center, signed the petition without hesitation.

Dr. West agreed with Dr. Goodman that the 10-year ABIM recertification is sufficient. He also denounced the ABIM testing process, costs, and content, saying he found the questions “so ambiguous that even knowing everything about the subject, I found the assignment of the ‘best’ answer to be a Talmudic interpretation.”

“The questions seem to have a sadistic level of complexity and ambiguity baked into them, rather than being a direct assessment of knowledge,” he explained.

The ABIM is also “completely opaque” about their process of developing questions and defining answers, Dr. West said. And “the ABIM offers no data to support that their processes improve any clinical outcomes.”

Yet, the MOC “forces physicians to spend several hundred dollars every year as well as an incredible amount of their time jumping through hoops at the behest of ABIM to satisfy these imposed requirements,” Dr. West said. The time spent satisfying these requirements is also “strip-mining physician morale” by taking time away from families and personal lives.

As for ABIM finances, Dr. West said the organization offers no justification for “the extortionate costs imposed by this de facto monopoly.”

The glimpses we do see, however, “indicate an organization spending on a lavish condominium and offering conspicuously generous remuneration to its own leadership. The only thing that is assured by the ABIM’s MOC program is that it is wildly profitable for the ABIM,” he added.

In a tweet, he called on physicians to take a stand: “If you think MOC is good, say it. Otherwise, if you don’t sign [the petition], ask yourself why you don’t have the courage & character to do so.”
 

 

 

Next steps?

Dr. Goodman’s petition lays out potential alternatives to the MOC that “would better support physician competence and continuing education without imposing unnecessary hurdles.”

Alternatives include encouraging voluntary, accessible, and evidence-based CME programs to promote lifelong learning among physicians, establishing a system for peer evaluation and feedback, encouraging self-assessment, and fostering a culture of continuous quality improvement.

Dr. Goodman’s goal is to garner at least 10,000 signatures and reach out to credentialing committees around the country with the results to promote alternatives to MOC. As of the morning of August 1, the petition had more than 7,900 signatures.

He also gives Dr. Baron credit for his willingness to have a conversation about the MOC and intends for that conversation to be a civil, respectful debate.

“I can’t think of anything he could say that will convince me [MOC] is the right thing to do, but we’ll see what he has to say,” Dr. Goodman said.

A version of this article appeared on Medscape.com.

Publications
Topics
Sections

A petition demanding an end to maintenance of certification (MOC) requirements from the American Board of Internal Medicine (ABIM) gained traction among oncologists in late July, garnering nearly 7,500 signatures in 10 days.

The MOC program, “originally intended to uphold the standards of medical practice and promote lifelong learning, has evolved into a complex and time-consuming process that poses significant challenges to practicing physicians,” according to the petition launched on July 21 by hematologist-oncologist Aaron Goodman, MD. The MOC “has become burdensome, costly, and lacks evidence to support its effectiveness in improving patient care or physician competence.”

Dr. Goodman, assistant professor at the University of California, San Diego, is scheduled to debate the matter with ABIM President and Chief Executive Officer Richard J. Baron, MD, during a Healthcare Unfiltered podcast recorded and hosted by Chadi Nabhan, MD. In the August 3 podcast, Dr. Goodman and Dr. Baron will respond to questions posed via tweets, Dr. Goodman said.

Twitter survey posted by Dr. Nabhan in advance of the debate asked physicians whether the cost of the MOC, the time required for testing, or data sharing by ABIM is most bothersome. Of 158 respondents, 71% selected “all of the above.”
 

ABIM touts MOC ‘values’  

The ABIM requires an initial certification assessment that costs thousands of dollars and must be repeated every 10 years. The annual MOC requirements, which were tacked on within the last decade, involve tests that cost $220 for the first certificate a physician holds and about $120 for each subsequent one.

Over the course of his career, Dr. Goodman estimates he will spend over $40,000 to maintain his three ABIM boards in medicine, hematology, and oncology.

The ABIM did not immediately respond to a request for comment on the petition and debate, but a page on the ABIM website touts the “values of MOC” and says there is “compelling evidence” that the MOC improves the value of care without sacrificing quality and that board-certified physicians earn more.

According to the website, the MOC program “provides doctors with a pathway to know that they are staying current in the medical knowledge they use to treat patients and make important care decisions daily.” The ABIM also says that the “program has evolved to include new assessment options and an increased recognition of the work doctors do every day” and that the ABIM “continuously collaborates with doctors to increase the relevance of exams.”

Aniruddha Singh, MD, also weighed in on the value of MOC in a July 13 ABIM blog post. Dr. Singh is a member of this year’s Cardiovascular Disease Traditional, 10-Year MOC Exam Approval Committee and program director for the General Cardiovascular Fellowship at Drexel University College of Medicine, Reading, Pa.

In his post, Dr. Singh states that the MOC “facilitates a broader perspective on a range of topics [and] enables me to delve deeper into relevant areas, fostering a comprehensive understanding that enhances my quality of care.”
 

Growing resistance

Although Dr. Goodman acknowledged the merit of board testing every decade or so, physicians already do continuing medical education (CME) to keep up-to-date on their specialties. Dr. Goodman believes that most physicians, other than those who work for ABIM, would agree that MOC is a waste of time and money.

“It’s a pain-in-the-ass module that you sit at home and Google – it’s not really any sort of assessment,” nor does it help protect the public, said Goodman. The MOC is ultimately “just a money grab.”

According to the ABIM’s website, the nonprofit has net assets of more than $73.2 million as of June 30, 2022. Last year, the ABIM’s revenue hit $71.9 million, with more than half coming from MOC fees and 48% from certification.

The ABIM also says it spent $58 million in 2022. A breakdown shows about 63% of that money went to administering (28%), researching (13%), overseeing (4%), and developing (18%) the certification and MOC exams and program. ABIM’s CEO Dr. Baron makes about $1.2 million a year, according to recent tax filings. The COO makes about $550,000 from the ABIM and “related” organizations.

Fed up with the requirements and cost, Dr. Goodman decided to launch the petition to see if others agreed and how many.

His petition, addressed to the ABIM, expresses “deep dissatisfaction” with the ABIM MOC program and “respectfully request[s] that the ABIM take immediate action to eliminate the MOC program and adopt alternative, less burdensome methods of ensuring physician competence and continuous professional development.” Dr. Goodman, alongside Vinay Prasad, MD, a hematologist-oncologist and professor at the University of California, San Francisco, reiterated these points in a piece highlighting the petition.

Shortly before the petition went public but after he had been vocal on Twitter about issues with the MOC, Dr. Goodman said he received an invitation to join the ABIM Board of Governors. “My hypothesis is they are trying to ‘friend’ me, so I get credentialed, and they get me to stop yelling. It just made me more pissed off. I don’t want to be any part of that,” he said.

H. Jack West, MD, a thoracic oncologist and associate professor at City of Hope Comprehensive Cancer Center, signed the petition without hesitation.

Dr. West agreed with Dr. Goodman that the 10-year ABIM recertification is sufficient. He also denounced the ABIM testing process, costs, and content, saying he found the questions “so ambiguous that even knowing everything about the subject, I found the assignment of the ‘best’ answer to be a Talmudic interpretation.”

“The questions seem to have a sadistic level of complexity and ambiguity baked into them, rather than being a direct assessment of knowledge,” he explained.

The ABIM is also “completely opaque” about their process of developing questions and defining answers, Dr. West said. And “the ABIM offers no data to support that their processes improve any clinical outcomes.”

Yet, the MOC “forces physicians to spend several hundred dollars every year as well as an incredible amount of their time jumping through hoops at the behest of ABIM to satisfy these imposed requirements,” Dr. West said. The time spent satisfying these requirements is also “strip-mining physician morale” by taking time away from families and personal lives.

As for ABIM finances, Dr. West said the organization offers no justification for “the extortionate costs imposed by this de facto monopoly.”

The glimpses we do see, however, “indicate an organization spending on a lavish condominium and offering conspicuously generous remuneration to its own leadership. The only thing that is assured by the ABIM’s MOC program is that it is wildly profitable for the ABIM,” he added.

In a tweet, he called on physicians to take a stand: “If you think MOC is good, say it. Otherwise, if you don’t sign [the petition], ask yourself why you don’t have the courage & character to do so.”
 

 

 

Next steps?

Dr. Goodman’s petition lays out potential alternatives to the MOC that “would better support physician competence and continuing education without imposing unnecessary hurdles.”

Alternatives include encouraging voluntary, accessible, and evidence-based CME programs to promote lifelong learning among physicians, establishing a system for peer evaluation and feedback, encouraging self-assessment, and fostering a culture of continuous quality improvement.

Dr. Goodman’s goal is to garner at least 10,000 signatures and reach out to credentialing committees around the country with the results to promote alternatives to MOC. As of the morning of August 1, the petition had more than 7,900 signatures.

He also gives Dr. Baron credit for his willingness to have a conversation about the MOC and intends for that conversation to be a civil, respectful debate.

“I can’t think of anything he could say that will convince me [MOC] is the right thing to do, but we’ll see what he has to say,” Dr. Goodman said.

A version of this article appeared on Medscape.com.

A petition demanding an end to maintenance of certification (MOC) requirements from the American Board of Internal Medicine (ABIM) gained traction among oncologists in late July, garnering nearly 7,500 signatures in 10 days.

The MOC program, “originally intended to uphold the standards of medical practice and promote lifelong learning, has evolved into a complex and time-consuming process that poses significant challenges to practicing physicians,” according to the petition launched on July 21 by hematologist-oncologist Aaron Goodman, MD. The MOC “has become burdensome, costly, and lacks evidence to support its effectiveness in improving patient care or physician competence.”

Dr. Goodman, assistant professor at the University of California, San Diego, is scheduled to debate the matter with ABIM President and Chief Executive Officer Richard J. Baron, MD, during a Healthcare Unfiltered podcast recorded and hosted by Chadi Nabhan, MD. In the August 3 podcast, Dr. Goodman and Dr. Baron will respond to questions posed via tweets, Dr. Goodman said.

Twitter survey posted by Dr. Nabhan in advance of the debate asked physicians whether the cost of the MOC, the time required for testing, or data sharing by ABIM is most bothersome. Of 158 respondents, 71% selected “all of the above.”
 

ABIM touts MOC ‘values’  

The ABIM requires an initial certification assessment that costs thousands of dollars and must be repeated every 10 years. The annual MOC requirements, which were tacked on within the last decade, involve tests that cost $220 for the first certificate a physician holds and about $120 for each subsequent one.

Over the course of his career, Dr. Goodman estimates he will spend over $40,000 to maintain his three ABIM boards in medicine, hematology, and oncology.

The ABIM did not immediately respond to a request for comment on the petition and debate, but a page on the ABIM website touts the “values of MOC” and says there is “compelling evidence” that the MOC improves the value of care without sacrificing quality and that board-certified physicians earn more.

According to the website, the MOC program “provides doctors with a pathway to know that they are staying current in the medical knowledge they use to treat patients and make important care decisions daily.” The ABIM also says that the “program has evolved to include new assessment options and an increased recognition of the work doctors do every day” and that the ABIM “continuously collaborates with doctors to increase the relevance of exams.”

Aniruddha Singh, MD, also weighed in on the value of MOC in a July 13 ABIM blog post. Dr. Singh is a member of this year’s Cardiovascular Disease Traditional, 10-Year MOC Exam Approval Committee and program director for the General Cardiovascular Fellowship at Drexel University College of Medicine, Reading, Pa.

In his post, Dr. Singh states that the MOC “facilitates a broader perspective on a range of topics [and] enables me to delve deeper into relevant areas, fostering a comprehensive understanding that enhances my quality of care.”
 

Growing resistance

Although Dr. Goodman acknowledged the merit of board testing every decade or so, physicians already do continuing medical education (CME) to keep up-to-date on their specialties. Dr. Goodman believes that most physicians, other than those who work for ABIM, would agree that MOC is a waste of time and money.

“It’s a pain-in-the-ass module that you sit at home and Google – it’s not really any sort of assessment,” nor does it help protect the public, said Goodman. The MOC is ultimately “just a money grab.”

According to the ABIM’s website, the nonprofit has net assets of more than $73.2 million as of June 30, 2022. Last year, the ABIM’s revenue hit $71.9 million, with more than half coming from MOC fees and 48% from certification.

The ABIM also says it spent $58 million in 2022. A breakdown shows about 63% of that money went to administering (28%), researching (13%), overseeing (4%), and developing (18%) the certification and MOC exams and program. ABIM’s CEO Dr. Baron makes about $1.2 million a year, according to recent tax filings. The COO makes about $550,000 from the ABIM and “related” organizations.

Fed up with the requirements and cost, Dr. Goodman decided to launch the petition to see if others agreed and how many.

His petition, addressed to the ABIM, expresses “deep dissatisfaction” with the ABIM MOC program and “respectfully request[s] that the ABIM take immediate action to eliminate the MOC program and adopt alternative, less burdensome methods of ensuring physician competence and continuous professional development.” Dr. Goodman, alongside Vinay Prasad, MD, a hematologist-oncologist and professor at the University of California, San Francisco, reiterated these points in a piece highlighting the petition.

Shortly before the petition went public but after he had been vocal on Twitter about issues with the MOC, Dr. Goodman said he received an invitation to join the ABIM Board of Governors. “My hypothesis is they are trying to ‘friend’ me, so I get credentialed, and they get me to stop yelling. It just made me more pissed off. I don’t want to be any part of that,” he said.

H. Jack West, MD, a thoracic oncologist and associate professor at City of Hope Comprehensive Cancer Center, signed the petition without hesitation.

Dr. West agreed with Dr. Goodman that the 10-year ABIM recertification is sufficient. He also denounced the ABIM testing process, costs, and content, saying he found the questions “so ambiguous that even knowing everything about the subject, I found the assignment of the ‘best’ answer to be a Talmudic interpretation.”

“The questions seem to have a sadistic level of complexity and ambiguity baked into them, rather than being a direct assessment of knowledge,” he explained.

The ABIM is also “completely opaque” about their process of developing questions and defining answers, Dr. West said. And “the ABIM offers no data to support that their processes improve any clinical outcomes.”

Yet, the MOC “forces physicians to spend several hundred dollars every year as well as an incredible amount of their time jumping through hoops at the behest of ABIM to satisfy these imposed requirements,” Dr. West said. The time spent satisfying these requirements is also “strip-mining physician morale” by taking time away from families and personal lives.

As for ABIM finances, Dr. West said the organization offers no justification for “the extortionate costs imposed by this de facto monopoly.”

The glimpses we do see, however, “indicate an organization spending on a lavish condominium and offering conspicuously generous remuneration to its own leadership. The only thing that is assured by the ABIM’s MOC program is that it is wildly profitable for the ABIM,” he added.

In a tweet, he called on physicians to take a stand: “If you think MOC is good, say it. Otherwise, if you don’t sign [the petition], ask yourself why you don’t have the courage & character to do so.”
 

 

 

Next steps?

Dr. Goodman’s petition lays out potential alternatives to the MOC that “would better support physician competence and continuing education without imposing unnecessary hurdles.”

Alternatives include encouraging voluntary, accessible, and evidence-based CME programs to promote lifelong learning among physicians, establishing a system for peer evaluation and feedback, encouraging self-assessment, and fostering a culture of continuous quality improvement.

Dr. Goodman’s goal is to garner at least 10,000 signatures and reach out to credentialing committees around the country with the results to promote alternatives to MOC. As of the morning of August 1, the petition had more than 7,900 signatures.

He also gives Dr. Baron credit for his willingness to have a conversation about the MOC and intends for that conversation to be a civil, respectful debate.

“I can’t think of anything he could say that will convince me [MOC] is the right thing to do, but we’ll see what he has to say,” Dr. Goodman said.

A version of this article appeared on Medscape.com.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

New global initiative aims to reform cancer trials and care

Article Type
Changed
Mon, 09/11/2023 - 10:59

After 15 years of researching what works well in oncology – and where the field has gone awry – Christopher Booth, MD, had a career moment.

“As I approached mid-career, I realized publishing and describing problems wasn’t fulfilling. It wasn’t doing enough,” recalled Dr. Booth, an oncologist and professor at Queen’s University, Kingston, Ont. “I wanted to change mindsets and change systems so that things actually improved for the better for patients.”

His colleague, Bishal Gyawali, MD, PhD, described a similar epiphany. As a trainee, he noticed that the real-world effects of some so-called blockbuster cancer drugs too often failed to measure up to the hype.

“I realized we were lacking common sense in oncology,” said Dr. Gyawali, a medical oncologist and assistant professor at Queen’s University.

In 2019, Dr. Gyawali launched a Medscape column addressing what he considers to be that lack of common sense, and in 2022, he and Dr. Booth published a similarly titled opinion piece in Nature Medicine. The core idea: The cancer community needs to prioritize cancer treatments that benefit patients, treatments that meaningfully improve survival and quality of life.

Aaron Goodman, MD, a hematologist and associate professor at UC San Diego Health, was on the same page. He’d been interested in the evidence-based medicine movement since his time as a hematology fellow when that movement was “a bit of a counterculture,” he explained.

Dr. Goodman and Dr. Booth connected through their common interests and collaborated on a 2021 paper exploring the discomfort clinicians might feel when a patient’s needs fall on the “edge of oncology”: that is, when the guideline-recommended standard of care offers marginal benefit, at best, and could, at worst, cause patient harm.

“We said, ‘Now is the time to make change,’ ” he recalled. It was time to stop talking and do something.
 

Common sense and a common purpose

Dr. Booth, Dr. Gyawali, and Dr. Goodman joined forces and, with the backing of a philanthropist who had experience as a patient with cancer, convened an organizing committee of more than 30 like-minded oncologists and patient advocates from across the globe.

The group convened for a 3-day “meeting of the minds” in Kingston in April and laid out their intentions in a position paper published online in The Lancet Oncology.

The publication marks the official launch of an ambitious, multipronged, global initiative to enact change: Common Sense Oncology, a new patient-centered movement in cancer care.

In their paper, the committee outline the vision for Common Sense Oncology. The mission: prioritize patient-centered and equitable care by focusing on treatments that improve survival and quality of life, communication that promotes informed decision-making, and systems that ensure access to all patients.

However, increasingly, the cancer community faces a “troubling paradox,” the team wrote in The Lancet. In some instance, treatments that bring minimal benefit are overused while those that can make a meaningful difference in patients’ lives are not accessible to most worldwide.

One reason for this shift: Commercial interests, rather than patient interests, appear to be driving cancer research and care. The team explained, for instance, that over the past few decades, clinical trials have largely pivoted from publicly funded efforts to industry funded ones “designed to achieve regulatory approval or commercial advantage, [often] at the expense of investigating new approaches to surgery, radiotherapy, palliative care, and prevention.”

But “patients deserve better,” the group wrote.

The team outlined three pillars for the initiative: evidence generation, evidence interpretation, and evidence communication.

The evidence generation pillar will aim to improve trial design and reporting to prioritize outcomes that matter to patients.

“One concern is that over the last 10 years or so, most of our new treatments have had very, very small benefits, and we think the bar has dropped too low,” Dr. Booth said, explaining that many trials have moved away from focusing on improving survival and quality of life and toward detecting small differences between treatments on other endpoints – namely progression-free survival. “Those small benefits need to be balanced against the very real risks to our patients.”

The evidence interpretation pillar will aim to foster critical thinking so that clinicians can better identify poorly designed or reported trials and help patients make more informed decisions.

Lastly, the evidence communication pillar will focus on fostering better communication about treatment options among patients, the public, and policymakers. Without clear and thoughtful communication, patients may have unrealistic expectations about the effectiveness of treatments that offer only marginal clinical benefits.

The team also emphasized a need to focus on improving global equity and access to affordable treatments so all patients can benefit from care that extends survival or quality of life.

It’s an ambitious undertaking, especially for a group of full-time clinicians, researchers, and patient advocates “volunteering their time for societal good,” said Dr. Gyawali, but the project teams intend to hit the ground running.

The team has established short-term targets, such as identifying deficiencies in data interpretation within education programs within 6 months and developing educational materials that begin to correct those deficiencies within 12 months, Dr. Booth explained. In the longer term, the team will also aim to design clinical trials that focus on patient outcomes, such as overall survival and quality of life.

Breast cancer survivor and patient advocate Michelle Tregear, PhD, who was recruited to help with Common Sense Oncology, also hopes the initiative will lead to better regulatory control that requires trial sponsors to “focus on what matters to patients, not on surrogate endpoints.”

When it comes to clinical trials, “more, more, more is not always better,” said Dr. Tregear, director of Education and Training Programs for patient advocates at the National Breast Cancer Coalition, Washington, D.C. “Industry interests are not always aligned with patient interests,” and “the system, by and large, is not addressing questions that really matter to patients and their families.”

Although “it’s a tall order to change the direction that we’re going in,” Dr. Tregear is up to the challenge of helping raise awareness, which will hopefully spur patients to demand change.

When Dr. Goodman announced the Common Sense Oncology initiative on Twitter, the news brought excitement, with many oncologists asking to join.

With its sweeping, ambitious goals, the Common Sense Oncology initiative has a long road ahead. Figuring out how to implement some of its aims in practice will take time, Dr. Booth acknowledges, and the initial launch marks the first steps, which will continue to evolve over time.

“We’re not proposing we have all the answers or that we know what every patient would want – we’re saying we’ve not done a good job of communicating to patients the relative benefits and risks of different treatments,” Dr. Booth explained. “We want to celebrate and promote what helps and speak out about what’s not in the best interest of patients.”

Dr. Goodman reported consulting fees from Seattle Genetics and speaking honoraria from Curio. Dr. Booth, Dr. Gyawali, and Dr. Tregear reported having no financial conflicts of interest.

A version of this article appeared on Medscape.com.

Publications
Topics
Sections

After 15 years of researching what works well in oncology – and where the field has gone awry – Christopher Booth, MD, had a career moment.

“As I approached mid-career, I realized publishing and describing problems wasn’t fulfilling. It wasn’t doing enough,” recalled Dr. Booth, an oncologist and professor at Queen’s University, Kingston, Ont. “I wanted to change mindsets and change systems so that things actually improved for the better for patients.”

His colleague, Bishal Gyawali, MD, PhD, described a similar epiphany. As a trainee, he noticed that the real-world effects of some so-called blockbuster cancer drugs too often failed to measure up to the hype.

“I realized we were lacking common sense in oncology,” said Dr. Gyawali, a medical oncologist and assistant professor at Queen’s University.

In 2019, Dr. Gyawali launched a Medscape column addressing what he considers to be that lack of common sense, and in 2022, he and Dr. Booth published a similarly titled opinion piece in Nature Medicine. The core idea: The cancer community needs to prioritize cancer treatments that benefit patients, treatments that meaningfully improve survival and quality of life.

Aaron Goodman, MD, a hematologist and associate professor at UC San Diego Health, was on the same page. He’d been interested in the evidence-based medicine movement since his time as a hematology fellow when that movement was “a bit of a counterculture,” he explained.

Dr. Goodman and Dr. Booth connected through their common interests and collaborated on a 2021 paper exploring the discomfort clinicians might feel when a patient’s needs fall on the “edge of oncology”: that is, when the guideline-recommended standard of care offers marginal benefit, at best, and could, at worst, cause patient harm.

“We said, ‘Now is the time to make change,’ ” he recalled. It was time to stop talking and do something.
 

Common sense and a common purpose

Dr. Booth, Dr. Gyawali, and Dr. Goodman joined forces and, with the backing of a philanthropist who had experience as a patient with cancer, convened an organizing committee of more than 30 like-minded oncologists and patient advocates from across the globe.

The group convened for a 3-day “meeting of the minds” in Kingston in April and laid out their intentions in a position paper published online in The Lancet Oncology.

The publication marks the official launch of an ambitious, multipronged, global initiative to enact change: Common Sense Oncology, a new patient-centered movement in cancer care.

In their paper, the committee outline the vision for Common Sense Oncology. The mission: prioritize patient-centered and equitable care by focusing on treatments that improve survival and quality of life, communication that promotes informed decision-making, and systems that ensure access to all patients.

However, increasingly, the cancer community faces a “troubling paradox,” the team wrote in The Lancet. In some instance, treatments that bring minimal benefit are overused while those that can make a meaningful difference in patients’ lives are not accessible to most worldwide.

One reason for this shift: Commercial interests, rather than patient interests, appear to be driving cancer research and care. The team explained, for instance, that over the past few decades, clinical trials have largely pivoted from publicly funded efforts to industry funded ones “designed to achieve regulatory approval or commercial advantage, [often] at the expense of investigating new approaches to surgery, radiotherapy, palliative care, and prevention.”

But “patients deserve better,” the group wrote.

The team outlined three pillars for the initiative: evidence generation, evidence interpretation, and evidence communication.

The evidence generation pillar will aim to improve trial design and reporting to prioritize outcomes that matter to patients.

“One concern is that over the last 10 years or so, most of our new treatments have had very, very small benefits, and we think the bar has dropped too low,” Dr. Booth said, explaining that many trials have moved away from focusing on improving survival and quality of life and toward detecting small differences between treatments on other endpoints – namely progression-free survival. “Those small benefits need to be balanced against the very real risks to our patients.”

The evidence interpretation pillar will aim to foster critical thinking so that clinicians can better identify poorly designed or reported trials and help patients make more informed decisions.

Lastly, the evidence communication pillar will focus on fostering better communication about treatment options among patients, the public, and policymakers. Without clear and thoughtful communication, patients may have unrealistic expectations about the effectiveness of treatments that offer only marginal clinical benefits.

The team also emphasized a need to focus on improving global equity and access to affordable treatments so all patients can benefit from care that extends survival or quality of life.

It’s an ambitious undertaking, especially for a group of full-time clinicians, researchers, and patient advocates “volunteering their time for societal good,” said Dr. Gyawali, but the project teams intend to hit the ground running.

The team has established short-term targets, such as identifying deficiencies in data interpretation within education programs within 6 months and developing educational materials that begin to correct those deficiencies within 12 months, Dr. Booth explained. In the longer term, the team will also aim to design clinical trials that focus on patient outcomes, such as overall survival and quality of life.

Breast cancer survivor and patient advocate Michelle Tregear, PhD, who was recruited to help with Common Sense Oncology, also hopes the initiative will lead to better regulatory control that requires trial sponsors to “focus on what matters to patients, not on surrogate endpoints.”

When it comes to clinical trials, “more, more, more is not always better,” said Dr. Tregear, director of Education and Training Programs for patient advocates at the National Breast Cancer Coalition, Washington, D.C. “Industry interests are not always aligned with patient interests,” and “the system, by and large, is not addressing questions that really matter to patients and their families.”

Although “it’s a tall order to change the direction that we’re going in,” Dr. Tregear is up to the challenge of helping raise awareness, which will hopefully spur patients to demand change.

When Dr. Goodman announced the Common Sense Oncology initiative on Twitter, the news brought excitement, with many oncologists asking to join.

With its sweeping, ambitious goals, the Common Sense Oncology initiative has a long road ahead. Figuring out how to implement some of its aims in practice will take time, Dr. Booth acknowledges, and the initial launch marks the first steps, which will continue to evolve over time.

“We’re not proposing we have all the answers or that we know what every patient would want – we’re saying we’ve not done a good job of communicating to patients the relative benefits and risks of different treatments,” Dr. Booth explained. “We want to celebrate and promote what helps and speak out about what’s not in the best interest of patients.”

Dr. Goodman reported consulting fees from Seattle Genetics and speaking honoraria from Curio. Dr. Booth, Dr. Gyawali, and Dr. Tregear reported having no financial conflicts of interest.

A version of this article appeared on Medscape.com.

After 15 years of researching what works well in oncology – and where the field has gone awry – Christopher Booth, MD, had a career moment.

“As I approached mid-career, I realized publishing and describing problems wasn’t fulfilling. It wasn’t doing enough,” recalled Dr. Booth, an oncologist and professor at Queen’s University, Kingston, Ont. “I wanted to change mindsets and change systems so that things actually improved for the better for patients.”

His colleague, Bishal Gyawali, MD, PhD, described a similar epiphany. As a trainee, he noticed that the real-world effects of some so-called blockbuster cancer drugs too often failed to measure up to the hype.

“I realized we were lacking common sense in oncology,” said Dr. Gyawali, a medical oncologist and assistant professor at Queen’s University.

In 2019, Dr. Gyawali launched a Medscape column addressing what he considers to be that lack of common sense, and in 2022, he and Dr. Booth published a similarly titled opinion piece in Nature Medicine. The core idea: The cancer community needs to prioritize cancer treatments that benefit patients, treatments that meaningfully improve survival and quality of life.

Aaron Goodman, MD, a hematologist and associate professor at UC San Diego Health, was on the same page. He’d been interested in the evidence-based medicine movement since his time as a hematology fellow when that movement was “a bit of a counterculture,” he explained.

Dr. Goodman and Dr. Booth connected through their common interests and collaborated on a 2021 paper exploring the discomfort clinicians might feel when a patient’s needs fall on the “edge of oncology”: that is, when the guideline-recommended standard of care offers marginal benefit, at best, and could, at worst, cause patient harm.

“We said, ‘Now is the time to make change,’ ” he recalled. It was time to stop talking and do something.
 

Common sense and a common purpose

Dr. Booth, Dr. Gyawali, and Dr. Goodman joined forces and, with the backing of a philanthropist who had experience as a patient with cancer, convened an organizing committee of more than 30 like-minded oncologists and patient advocates from across the globe.

The group convened for a 3-day “meeting of the minds” in Kingston in April and laid out their intentions in a position paper published online in The Lancet Oncology.

The publication marks the official launch of an ambitious, multipronged, global initiative to enact change: Common Sense Oncology, a new patient-centered movement in cancer care.

In their paper, the committee outline the vision for Common Sense Oncology. The mission: prioritize patient-centered and equitable care by focusing on treatments that improve survival and quality of life, communication that promotes informed decision-making, and systems that ensure access to all patients.

However, increasingly, the cancer community faces a “troubling paradox,” the team wrote in The Lancet. In some instance, treatments that bring minimal benefit are overused while those that can make a meaningful difference in patients’ lives are not accessible to most worldwide.

One reason for this shift: Commercial interests, rather than patient interests, appear to be driving cancer research and care. The team explained, for instance, that over the past few decades, clinical trials have largely pivoted from publicly funded efforts to industry funded ones “designed to achieve regulatory approval or commercial advantage, [often] at the expense of investigating new approaches to surgery, radiotherapy, palliative care, and prevention.”

But “patients deserve better,” the group wrote.

The team outlined three pillars for the initiative: evidence generation, evidence interpretation, and evidence communication.

The evidence generation pillar will aim to improve trial design and reporting to prioritize outcomes that matter to patients.

“One concern is that over the last 10 years or so, most of our new treatments have had very, very small benefits, and we think the bar has dropped too low,” Dr. Booth said, explaining that many trials have moved away from focusing on improving survival and quality of life and toward detecting small differences between treatments on other endpoints – namely progression-free survival. “Those small benefits need to be balanced against the very real risks to our patients.”

The evidence interpretation pillar will aim to foster critical thinking so that clinicians can better identify poorly designed or reported trials and help patients make more informed decisions.

Lastly, the evidence communication pillar will focus on fostering better communication about treatment options among patients, the public, and policymakers. Without clear and thoughtful communication, patients may have unrealistic expectations about the effectiveness of treatments that offer only marginal clinical benefits.

The team also emphasized a need to focus on improving global equity and access to affordable treatments so all patients can benefit from care that extends survival or quality of life.

It’s an ambitious undertaking, especially for a group of full-time clinicians, researchers, and patient advocates “volunteering their time for societal good,” said Dr. Gyawali, but the project teams intend to hit the ground running.

The team has established short-term targets, such as identifying deficiencies in data interpretation within education programs within 6 months and developing educational materials that begin to correct those deficiencies within 12 months, Dr. Booth explained. In the longer term, the team will also aim to design clinical trials that focus on patient outcomes, such as overall survival and quality of life.

Breast cancer survivor and patient advocate Michelle Tregear, PhD, who was recruited to help with Common Sense Oncology, also hopes the initiative will lead to better regulatory control that requires trial sponsors to “focus on what matters to patients, not on surrogate endpoints.”

When it comes to clinical trials, “more, more, more is not always better,” said Dr. Tregear, director of Education and Training Programs for patient advocates at the National Breast Cancer Coalition, Washington, D.C. “Industry interests are not always aligned with patient interests,” and “the system, by and large, is not addressing questions that really matter to patients and their families.”

Although “it’s a tall order to change the direction that we’re going in,” Dr. Tregear is up to the challenge of helping raise awareness, which will hopefully spur patients to demand change.

When Dr. Goodman announced the Common Sense Oncology initiative on Twitter, the news brought excitement, with many oncologists asking to join.

With its sweeping, ambitious goals, the Common Sense Oncology initiative has a long road ahead. Figuring out how to implement some of its aims in practice will take time, Dr. Booth acknowledges, and the initial launch marks the first steps, which will continue to evolve over time.

“We’re not proposing we have all the answers or that we know what every patient would want – we’re saying we’ve not done a good job of communicating to patients the relative benefits and risks of different treatments,” Dr. Booth explained. “We want to celebrate and promote what helps and speak out about what’s not in the best interest of patients.”

Dr. Goodman reported consulting fees from Seattle Genetics and speaking honoraria from Curio. Dr. Booth, Dr. Gyawali, and Dr. Tregear reported having no financial conflicts of interest.

A version of this article appeared on Medscape.com.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

WHO declares aspartame as possible carcinogen

Article Type
Changed
Fri, 07/14/2023 - 12:13

The International Agency for Research on Cancer announced on July 13 that it has officially classified the artificial sweetener aspartame as a possible carcinogen.

Although this means aspartame might cause cancer in humans, the Group 2B classification from the IARC means the evidence is “limited.” A summary of the working group’s evaluation, also published in The Lancet Oncology, explained that the classification was based on data from three studies assessing the link between aspartame intake and primary liver cancer.

Using that evidence, the World Health Organization and Food and Agriculture Organization Joint Expert Committee on Food Additives confirmed its existing stance that aspartame consumption of up to 40 mg/kg of body weight per day – the amount found in 9-14 diet soft drinks – is safe.

The decision, which was anticipated by Reuters in late June, drew praise from an array of experts who weighed in on the study results via the U.K.-based Science Media Centre. Many emphasized the lack of data showing a causal relationship between the low-calorie artificial sweetener and sought to temper any alarmism related to the decision.

“In short the evidence that aspartame causes primary liver cancer, or any other cancer in humans, is very weak,” said Paul Pharoah, MD, PhD, a professor of cancer epidemiology at Cedars-Sinai Medical Center, Los Angeles. “Group 2B is a very conservative classification in that almost any evidence of carcinogenicity, however flawed, will put a chemical in that category or above.”

Other examples of substances classified as Group 2B are extract of aloe vera, diesel oil, and caffeic acid found in coffee and tea, Dr. Pharoah explained, adding that “this is reflected in the view of the [JECFA] who concluded that there was no convincing evidence from experimental animal or human data that aspartame has adverse effects after ingestion.”

“The general public should not be worried about the risk of cancer associated with a chemical classed as Group 2B by IARC,” he stressed.

Alan Boobis, OBE, PhD, similarly noted that the Group 2B classification “reflects a lack of confidence that the data from experimental animals or from humans is sufficiently convincing to reach a clear conclusion that aspartame is carcinogenic.”

“Hence, exposure at current levels would not be anticipated to have any detrimental effects,” added Dr. Boobis, emeritus professor of toxicology, Imperial College London.

The IARC/JECFA opinion is “very welcome” and “ends the speculation about the safety of aspartame,” added Gunter Kuhnle, Dr. rer. nat., a professor of nutrition and food science at the University of Reading (England).

“It is unfortunate that leaking some information might have created unnecessary uncertainty and concern as consumers might be rightfully worried if they are told that something that is in many foods could cause cancer,” Dr. Kuhnle said. “The published opinion puts this into perspective and makes it very clear that there is no cause for concern when consumed at the current amounts.”

The data reviewed by the IARC Working Group included three studies, comprising four prospective cohorts, that “assessed the association of artificially sweetened beverage consumption with liver cancer risk,” the group reported.

The cohort studies – including one conducted within 10 European countries, one that pooled data from two large U.S. cohorts, and a prospective study also conducted in the United States – each “showed positive associations between artificially sweetened beverage consumption and cancer incidence or cancer mortality” in the overall study population or in relevant subgroups.

Although the studies were of “high quality and controlled for many potential confounders,” the Working Group concluded that “chance, bias, or confounding could not be ruled out with reasonable confidence.” Thus, the evidence for cancer in humans was deemed “limited” for hepatocellular carcinoma and “inadequate” for other cancer types.”

Dr. Pharoah and Dr. Kuhnle disclosed no relevant financial relationships. Dr. Boobis is a member of a number of advisory committees in the public and private sectors, including the International Life Science Institute and The Center for Research on Ingredient Safety at Michigan State University.

A version of this article first appeared on Medscape.com.

Publications
Topics
Sections

The International Agency for Research on Cancer announced on July 13 that it has officially classified the artificial sweetener aspartame as a possible carcinogen.

Although this means aspartame might cause cancer in humans, the Group 2B classification from the IARC means the evidence is “limited.” A summary of the working group’s evaluation, also published in The Lancet Oncology, explained that the classification was based on data from three studies assessing the link between aspartame intake and primary liver cancer.

Using that evidence, the World Health Organization and Food and Agriculture Organization Joint Expert Committee on Food Additives confirmed its existing stance that aspartame consumption of up to 40 mg/kg of body weight per day – the amount found in 9-14 diet soft drinks – is safe.

The decision, which was anticipated by Reuters in late June, drew praise from an array of experts who weighed in on the study results via the U.K.-based Science Media Centre. Many emphasized the lack of data showing a causal relationship between the low-calorie artificial sweetener and sought to temper any alarmism related to the decision.

“In short the evidence that aspartame causes primary liver cancer, or any other cancer in humans, is very weak,” said Paul Pharoah, MD, PhD, a professor of cancer epidemiology at Cedars-Sinai Medical Center, Los Angeles. “Group 2B is a very conservative classification in that almost any evidence of carcinogenicity, however flawed, will put a chemical in that category or above.”

Other examples of substances classified as Group 2B are extract of aloe vera, diesel oil, and caffeic acid found in coffee and tea, Dr. Pharoah explained, adding that “this is reflected in the view of the [JECFA] who concluded that there was no convincing evidence from experimental animal or human data that aspartame has adverse effects after ingestion.”

“The general public should not be worried about the risk of cancer associated with a chemical classed as Group 2B by IARC,” he stressed.

Alan Boobis, OBE, PhD, similarly noted that the Group 2B classification “reflects a lack of confidence that the data from experimental animals or from humans is sufficiently convincing to reach a clear conclusion that aspartame is carcinogenic.”

“Hence, exposure at current levels would not be anticipated to have any detrimental effects,” added Dr. Boobis, emeritus professor of toxicology, Imperial College London.

The IARC/JECFA opinion is “very welcome” and “ends the speculation about the safety of aspartame,” added Gunter Kuhnle, Dr. rer. nat., a professor of nutrition and food science at the University of Reading (England).

“It is unfortunate that leaking some information might have created unnecessary uncertainty and concern as consumers might be rightfully worried if they are told that something that is in many foods could cause cancer,” Dr. Kuhnle said. “The published opinion puts this into perspective and makes it very clear that there is no cause for concern when consumed at the current amounts.”

The data reviewed by the IARC Working Group included three studies, comprising four prospective cohorts, that “assessed the association of artificially sweetened beverage consumption with liver cancer risk,” the group reported.

The cohort studies – including one conducted within 10 European countries, one that pooled data from two large U.S. cohorts, and a prospective study also conducted in the United States – each “showed positive associations between artificially sweetened beverage consumption and cancer incidence or cancer mortality” in the overall study population or in relevant subgroups.

Although the studies were of “high quality and controlled for many potential confounders,” the Working Group concluded that “chance, bias, or confounding could not be ruled out with reasonable confidence.” Thus, the evidence for cancer in humans was deemed “limited” for hepatocellular carcinoma and “inadequate” for other cancer types.”

Dr. Pharoah and Dr. Kuhnle disclosed no relevant financial relationships. Dr. Boobis is a member of a number of advisory committees in the public and private sectors, including the International Life Science Institute and The Center for Research on Ingredient Safety at Michigan State University.

A version of this article first appeared on Medscape.com.

The International Agency for Research on Cancer announced on July 13 that it has officially classified the artificial sweetener aspartame as a possible carcinogen.

Although this means aspartame might cause cancer in humans, the Group 2B classification from the IARC means the evidence is “limited.” A summary of the working group’s evaluation, also published in The Lancet Oncology, explained that the classification was based on data from three studies assessing the link between aspartame intake and primary liver cancer.

Using that evidence, the World Health Organization and Food and Agriculture Organization Joint Expert Committee on Food Additives confirmed its existing stance that aspartame consumption of up to 40 mg/kg of body weight per day – the amount found in 9-14 diet soft drinks – is safe.

The decision, which was anticipated by Reuters in late June, drew praise from an array of experts who weighed in on the study results via the U.K.-based Science Media Centre. Many emphasized the lack of data showing a causal relationship between the low-calorie artificial sweetener and sought to temper any alarmism related to the decision.

“In short the evidence that aspartame causes primary liver cancer, or any other cancer in humans, is very weak,” said Paul Pharoah, MD, PhD, a professor of cancer epidemiology at Cedars-Sinai Medical Center, Los Angeles. “Group 2B is a very conservative classification in that almost any evidence of carcinogenicity, however flawed, will put a chemical in that category or above.”

Other examples of substances classified as Group 2B are extract of aloe vera, diesel oil, and caffeic acid found in coffee and tea, Dr. Pharoah explained, adding that “this is reflected in the view of the [JECFA] who concluded that there was no convincing evidence from experimental animal or human data that aspartame has adverse effects after ingestion.”

“The general public should not be worried about the risk of cancer associated with a chemical classed as Group 2B by IARC,” he stressed.

Alan Boobis, OBE, PhD, similarly noted that the Group 2B classification “reflects a lack of confidence that the data from experimental animals or from humans is sufficiently convincing to reach a clear conclusion that aspartame is carcinogenic.”

“Hence, exposure at current levels would not be anticipated to have any detrimental effects,” added Dr. Boobis, emeritus professor of toxicology, Imperial College London.

The IARC/JECFA opinion is “very welcome” and “ends the speculation about the safety of aspartame,” added Gunter Kuhnle, Dr. rer. nat., a professor of nutrition and food science at the University of Reading (England).

“It is unfortunate that leaking some information might have created unnecessary uncertainty and concern as consumers might be rightfully worried if they are told that something that is in many foods could cause cancer,” Dr. Kuhnle said. “The published opinion puts this into perspective and makes it very clear that there is no cause for concern when consumed at the current amounts.”

The data reviewed by the IARC Working Group included three studies, comprising four prospective cohorts, that “assessed the association of artificially sweetened beverage consumption with liver cancer risk,” the group reported.

The cohort studies – including one conducted within 10 European countries, one that pooled data from two large U.S. cohorts, and a prospective study also conducted in the United States – each “showed positive associations between artificially sweetened beverage consumption and cancer incidence or cancer mortality” in the overall study population or in relevant subgroups.

Although the studies were of “high quality and controlled for many potential confounders,” the Working Group concluded that “chance, bias, or confounding could not be ruled out with reasonable confidence.” Thus, the evidence for cancer in humans was deemed “limited” for hepatocellular carcinoma and “inadequate” for other cancer types.”

Dr. Pharoah and Dr. Kuhnle disclosed no relevant financial relationships. Dr. Boobis is a member of a number of advisory committees in the public and private sectors, including the International Life Science Institute and The Center for Research on Ingredient Safety at Michigan State University.

A version of this article first appeared on Medscape.com.

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM THE LANCET ONCOLOGY

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Higher alcohol consumption linked to early-onset CRC

Article Type
Changed
Fri, 07/14/2023 - 09:54

 

TOPLINE:

Higher levels of alcohol consumption appear to increase an individual’s risk of early-onset colorectal cancer (CRC), particularly distal colon and rectal cancers, according to a population-based study from South Korea.

METHODOLOGY:

  • The investigators retrospectively compared average daily alcohol consumption with early-onset CRC risk among nearly 5.7 million adults younger than 50 years, using data from the Korean National Health Insurance Service.
  • Alcohol consumption levels were defined as nondrinker, light (< 10 g/day or < 0.7 U.S. drinks/day), moderate (10-30 g/day for men, 10-20 g/day for women), and heavy (≥ 30 g/day or ≥ 2.1 drinks/day for men, ≥ 20 g/day or ≥ 1.4 drinks/day for women).
  • The primary outcome was incidence of early-onset CRC diagnosed before age 50. Models were adjusted for age, sex, smoking status, exercise, and income, as well as for comorbidities.

TAKEAWAY:

  • Overall, 8,314 incident early-onset CRC cases occurred during the mean follow-up period of 7.4 years.
  • Compared with light drinking, moderate and heavy drinking were associated with a significantly elevated risk of early-onset CRC (adjusted hazard ratio, 1.09 and 1.20, respectively); by sex, significant associations were found only among men.
  • Among men, heavy drinking vs. light drinking was associated with a 26% increased risk of distal colon cancer, a 17% higher risk of rectal cancer, and a 29% higher risk of unspecified colon cancer (but not proximal colon cancer).
  • Among women, moderate drinking was associated with a 47% increased risk of distal colon cancer. Among nondrinkers, there was a 14% reduced risk of rectal cancer, compared with light drinkers.

IN PRACTICE:

“This population-based study provides evidence that higher levels of alcohol consumption may increase the risk of early-onset CRC,” the investigators concluded. “[E]ffective interventions are required to discourage alcohol consumption among young people and to tailor CRC screening approaches for high-risk individuals.”

SOURCE:

The study was led by researchers at Seoul National University, South Korea. It was published online in the Journal of Clinical Oncology.

LIMITATIONS:

Study limitations include self-reported alcohol consumption. Data were missing for a higher number of male participants and younger participants, and there was a potential problem related to multiple comparisons and confounders. Only Korean individuals were included in the study, so larger studies involving various races are needed.

DISCLOSURES:

Funding was provided by grants from the Korea Health Technology R&D Project and the Ministry of Health and Welfare, Republic of Korea. No potential conflicts of interest were reported.

A version of this article first appeared on Medscape.com.

Publications
Topics
Sections

 

TOPLINE:

Higher levels of alcohol consumption appear to increase an individual’s risk of early-onset colorectal cancer (CRC), particularly distal colon and rectal cancers, according to a population-based study from South Korea.

METHODOLOGY:

  • The investigators retrospectively compared average daily alcohol consumption with early-onset CRC risk among nearly 5.7 million adults younger than 50 years, using data from the Korean National Health Insurance Service.
  • Alcohol consumption levels were defined as nondrinker, light (< 10 g/day or < 0.7 U.S. drinks/day), moderate (10-30 g/day for men, 10-20 g/day for women), and heavy (≥ 30 g/day or ≥ 2.1 drinks/day for men, ≥ 20 g/day or ≥ 1.4 drinks/day for women).
  • The primary outcome was incidence of early-onset CRC diagnosed before age 50. Models were adjusted for age, sex, smoking status, exercise, and income, as well as for comorbidities.

TAKEAWAY:

  • Overall, 8,314 incident early-onset CRC cases occurred during the mean follow-up period of 7.4 years.
  • Compared with light drinking, moderate and heavy drinking were associated with a significantly elevated risk of early-onset CRC (adjusted hazard ratio, 1.09 and 1.20, respectively); by sex, significant associations were found only among men.
  • Among men, heavy drinking vs. light drinking was associated with a 26% increased risk of distal colon cancer, a 17% higher risk of rectal cancer, and a 29% higher risk of unspecified colon cancer (but not proximal colon cancer).
  • Among women, moderate drinking was associated with a 47% increased risk of distal colon cancer. Among nondrinkers, there was a 14% reduced risk of rectal cancer, compared with light drinkers.

IN PRACTICE:

“This population-based study provides evidence that higher levels of alcohol consumption may increase the risk of early-onset CRC,” the investigators concluded. “[E]ffective interventions are required to discourage alcohol consumption among young people and to tailor CRC screening approaches for high-risk individuals.”

SOURCE:

The study was led by researchers at Seoul National University, South Korea. It was published online in the Journal of Clinical Oncology.

LIMITATIONS:

Study limitations include self-reported alcohol consumption. Data were missing for a higher number of male participants and younger participants, and there was a potential problem related to multiple comparisons and confounders. Only Korean individuals were included in the study, so larger studies involving various races are needed.

DISCLOSURES:

Funding was provided by grants from the Korea Health Technology R&D Project and the Ministry of Health and Welfare, Republic of Korea. No potential conflicts of interest were reported.

A version of this article first appeared on Medscape.com.

 

TOPLINE:

Higher levels of alcohol consumption appear to increase an individual’s risk of early-onset colorectal cancer (CRC), particularly distal colon and rectal cancers, according to a population-based study from South Korea.

METHODOLOGY:

  • The investigators retrospectively compared average daily alcohol consumption with early-onset CRC risk among nearly 5.7 million adults younger than 50 years, using data from the Korean National Health Insurance Service.
  • Alcohol consumption levels were defined as nondrinker, light (< 10 g/day or < 0.7 U.S. drinks/day), moderate (10-30 g/day for men, 10-20 g/day for women), and heavy (≥ 30 g/day or ≥ 2.1 drinks/day for men, ≥ 20 g/day or ≥ 1.4 drinks/day for women).
  • The primary outcome was incidence of early-onset CRC diagnosed before age 50. Models were adjusted for age, sex, smoking status, exercise, and income, as well as for comorbidities.

TAKEAWAY:

  • Overall, 8,314 incident early-onset CRC cases occurred during the mean follow-up period of 7.4 years.
  • Compared with light drinking, moderate and heavy drinking were associated with a significantly elevated risk of early-onset CRC (adjusted hazard ratio, 1.09 and 1.20, respectively); by sex, significant associations were found only among men.
  • Among men, heavy drinking vs. light drinking was associated with a 26% increased risk of distal colon cancer, a 17% higher risk of rectal cancer, and a 29% higher risk of unspecified colon cancer (but not proximal colon cancer).
  • Among women, moderate drinking was associated with a 47% increased risk of distal colon cancer. Among nondrinkers, there was a 14% reduced risk of rectal cancer, compared with light drinkers.

IN PRACTICE:

“This population-based study provides evidence that higher levels of alcohol consumption may increase the risk of early-onset CRC,” the investigators concluded. “[E]ffective interventions are required to discourage alcohol consumption among young people and to tailor CRC screening approaches for high-risk individuals.”

SOURCE:

The study was led by researchers at Seoul National University, South Korea. It was published online in the Journal of Clinical Oncology.

LIMITATIONS:

Study limitations include self-reported alcohol consumption. Data were missing for a higher number of male participants and younger participants, and there was a potential problem related to multiple comparisons and confounders. Only Korean individuals were included in the study, so larger studies involving various races are needed.

DISCLOSURES:

Funding was provided by grants from the Korea Health Technology R&D Project and the Ministry of Health and Welfare, Republic of Korea. No potential conflicts of interest were reported.

A version of this article first appeared on Medscape.com.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article