User login
Lung adverse effects in patients taking trastuzumab deruxtecan
although the benefit-to-risk relationship with use of the drug is still positive, say researchers who report a review of early clinical trials with the drug.
T-DXd is a monoclonal antibody that targets HER2. It is approved for use in HER2-positive breast, gastric, and lung cancers.
In the new study, investigators analyzed data from early clinical trials that involved patients with advanced cancers who had been heavily pretreated. They found an incidence of just over 15% for interstitial lung disease (ILD)/pneumonitis associated with the drug. Most patients (77.4%) had grade 1 or 2 ILD, but 2.2% of patients had grade 5 ILD.
“Interstitial lung disease is a known risk factor in patients treated with antibody conjugates for cancer,” commented lead author Charles Powell, MD, Icahn School of Medicine at Mount Sinai, New York. This adverse effect can lead to lung fibrosis and can become severe, life threatening, and even fatal, the authors warned.
The authors also discussed management of the event, which involves corticosteroids, and recommended that any patient who develops ILD of grade 3 or higher be hospitalized.
Close monitoring and proactive management may reduce the risk of ILD, they suggested.
Indeed, the incidence of this adverse effect was lower in a later phase 3 trial of the drug (10.5% in the DESTINY-Breast03 trial) and that the adverse events were less severe in this patient population (none of these events were of grade 4 or 5).
“Increased knowledge ... and implementation of ILD/pneumonitis monitoring, diagnosis, and management guidelines” may have resulted in this adverse effect being identified early and treated before it progressed, they commented.
ILD is highlighted in a boxed warning on the product label.
The study was published online in ESMO Open.
In their review, the investigators evaluated nine early-stage monotherapy clinical trials (phases 1 and 2) involving a total of 1,150 patients (breast cancer, 44.3%; gastric cancer, 25.6%; lung cancer, 17.7%; colorectal cancer, 9.3%, other cancers, 3.0%).
These patients had advanced cancer and had been heavily pretreated with a median of four prior lines of therapy. They received one or more doses of at least 5.4 mg/kg of T-DXd.
Nearly half of the cohort were treated for more than 6 months. A total of 276 potential ILD/pneumonitis events were sent for adjudication; of those, 85% were adjudicated as ILD/pneumonitis.
The overall incidence of adjudicated ILD/pneumonitis events was 15.4%; most were low-grade events. Some 87% of patients experienced their first ILD event within 12 months of treatment. The median time to experiencing an ILD/pneumonitis event was 5.4 months.
Some of the patients who developed grade 1 ILD/pneumonitis were treated and the adverse event resolved. These patients were then rechallenged with the drug. Only 3 of the 47 rechallenged patients experienced recurrence of ILD/pneumonitis, the authors noted.
“Rechallenge with T-DXd after complete resolution of grade 1 events is possible and warrants further investigation,” they commented. They cautioned, however, that rechallenge is not recommended for all patients, at least not for those with grade 2 or higher ILD/pneumonitis.
Overall, the authors concluded that the “benefit-risk of T-DXd treatment is positive,” but they warned that some patients may be at increased risk of developing ILD/pneumonitis
Baseline factors that increase the risk of developing an ILD/pneumonitis event include the following: being younger than 65 years, receiving a T-DXd dose of more than6.4 mg/kg, having a baseline oxygen saturation level of less than 95%, having moderate to severe renal impairment, and having lung comorbidities. In addition, patients who had initially been diagnosed with cancer more than 4 years before receiving the drug were at higher risk of developing ILD/pneumonitis.
“Using learnings from the early clinical trials experience, physician education and patient management protocols were revised and disseminated by the study sponsors [and] more recent trial data in earlier lines of therapy has demonstrated lower rates of ILD events, suggesting close monitoring and proactive management of ILD/pneumonitis is warranted for all patients,” Dr. Powell said in a statement.
The T-DXd clinical trials were sponsored by AstraZeneca and Daiichi Sankyo. Dr. Powell has received fees from Daiichi Sankyo, AstraZeneca, and Voluntis.
A version of this article first appeared on Medscape.com.
although the benefit-to-risk relationship with use of the drug is still positive, say researchers who report a review of early clinical trials with the drug.
T-DXd is a monoclonal antibody that targets HER2. It is approved for use in HER2-positive breast, gastric, and lung cancers.
In the new study, investigators analyzed data from early clinical trials that involved patients with advanced cancers who had been heavily pretreated. They found an incidence of just over 15% for interstitial lung disease (ILD)/pneumonitis associated with the drug. Most patients (77.4%) had grade 1 or 2 ILD, but 2.2% of patients had grade 5 ILD.
“Interstitial lung disease is a known risk factor in patients treated with antibody conjugates for cancer,” commented lead author Charles Powell, MD, Icahn School of Medicine at Mount Sinai, New York. This adverse effect can lead to lung fibrosis and can become severe, life threatening, and even fatal, the authors warned.
The authors also discussed management of the event, which involves corticosteroids, and recommended that any patient who develops ILD of grade 3 or higher be hospitalized.
Close monitoring and proactive management may reduce the risk of ILD, they suggested.
Indeed, the incidence of this adverse effect was lower in a later phase 3 trial of the drug (10.5% in the DESTINY-Breast03 trial) and that the adverse events were less severe in this patient population (none of these events were of grade 4 or 5).
“Increased knowledge ... and implementation of ILD/pneumonitis monitoring, diagnosis, and management guidelines” may have resulted in this adverse effect being identified early and treated before it progressed, they commented.
ILD is highlighted in a boxed warning on the product label.
The study was published online in ESMO Open.
In their review, the investigators evaluated nine early-stage monotherapy clinical trials (phases 1 and 2) involving a total of 1,150 patients (breast cancer, 44.3%; gastric cancer, 25.6%; lung cancer, 17.7%; colorectal cancer, 9.3%, other cancers, 3.0%).
These patients had advanced cancer and had been heavily pretreated with a median of four prior lines of therapy. They received one or more doses of at least 5.4 mg/kg of T-DXd.
Nearly half of the cohort were treated for more than 6 months. A total of 276 potential ILD/pneumonitis events were sent for adjudication; of those, 85% were adjudicated as ILD/pneumonitis.
The overall incidence of adjudicated ILD/pneumonitis events was 15.4%; most were low-grade events. Some 87% of patients experienced their first ILD event within 12 months of treatment. The median time to experiencing an ILD/pneumonitis event was 5.4 months.
Some of the patients who developed grade 1 ILD/pneumonitis were treated and the adverse event resolved. These patients were then rechallenged with the drug. Only 3 of the 47 rechallenged patients experienced recurrence of ILD/pneumonitis, the authors noted.
“Rechallenge with T-DXd after complete resolution of grade 1 events is possible and warrants further investigation,” they commented. They cautioned, however, that rechallenge is not recommended for all patients, at least not for those with grade 2 or higher ILD/pneumonitis.
Overall, the authors concluded that the “benefit-risk of T-DXd treatment is positive,” but they warned that some patients may be at increased risk of developing ILD/pneumonitis
Baseline factors that increase the risk of developing an ILD/pneumonitis event include the following: being younger than 65 years, receiving a T-DXd dose of more than6.4 mg/kg, having a baseline oxygen saturation level of less than 95%, having moderate to severe renal impairment, and having lung comorbidities. In addition, patients who had initially been diagnosed with cancer more than 4 years before receiving the drug were at higher risk of developing ILD/pneumonitis.
“Using learnings from the early clinical trials experience, physician education and patient management protocols were revised and disseminated by the study sponsors [and] more recent trial data in earlier lines of therapy has demonstrated lower rates of ILD events, suggesting close monitoring and proactive management of ILD/pneumonitis is warranted for all patients,” Dr. Powell said in a statement.
The T-DXd clinical trials were sponsored by AstraZeneca and Daiichi Sankyo. Dr. Powell has received fees from Daiichi Sankyo, AstraZeneca, and Voluntis.
A version of this article first appeared on Medscape.com.
although the benefit-to-risk relationship with use of the drug is still positive, say researchers who report a review of early clinical trials with the drug.
T-DXd is a monoclonal antibody that targets HER2. It is approved for use in HER2-positive breast, gastric, and lung cancers.
In the new study, investigators analyzed data from early clinical trials that involved patients with advanced cancers who had been heavily pretreated. They found an incidence of just over 15% for interstitial lung disease (ILD)/pneumonitis associated with the drug. Most patients (77.4%) had grade 1 or 2 ILD, but 2.2% of patients had grade 5 ILD.
“Interstitial lung disease is a known risk factor in patients treated with antibody conjugates for cancer,” commented lead author Charles Powell, MD, Icahn School of Medicine at Mount Sinai, New York. This adverse effect can lead to lung fibrosis and can become severe, life threatening, and even fatal, the authors warned.
The authors also discussed management of the event, which involves corticosteroids, and recommended that any patient who develops ILD of grade 3 or higher be hospitalized.
Close monitoring and proactive management may reduce the risk of ILD, they suggested.
Indeed, the incidence of this adverse effect was lower in a later phase 3 trial of the drug (10.5% in the DESTINY-Breast03 trial) and that the adverse events were less severe in this patient population (none of these events were of grade 4 or 5).
“Increased knowledge ... and implementation of ILD/pneumonitis monitoring, diagnosis, and management guidelines” may have resulted in this adverse effect being identified early and treated before it progressed, they commented.
ILD is highlighted in a boxed warning on the product label.
The study was published online in ESMO Open.
In their review, the investigators evaluated nine early-stage monotherapy clinical trials (phases 1 and 2) involving a total of 1,150 patients (breast cancer, 44.3%; gastric cancer, 25.6%; lung cancer, 17.7%; colorectal cancer, 9.3%, other cancers, 3.0%).
These patients had advanced cancer and had been heavily pretreated with a median of four prior lines of therapy. They received one or more doses of at least 5.4 mg/kg of T-DXd.
Nearly half of the cohort were treated for more than 6 months. A total of 276 potential ILD/pneumonitis events were sent for adjudication; of those, 85% were adjudicated as ILD/pneumonitis.
The overall incidence of adjudicated ILD/pneumonitis events was 15.4%; most were low-grade events. Some 87% of patients experienced their first ILD event within 12 months of treatment. The median time to experiencing an ILD/pneumonitis event was 5.4 months.
Some of the patients who developed grade 1 ILD/pneumonitis were treated and the adverse event resolved. These patients were then rechallenged with the drug. Only 3 of the 47 rechallenged patients experienced recurrence of ILD/pneumonitis, the authors noted.
“Rechallenge with T-DXd after complete resolution of grade 1 events is possible and warrants further investigation,” they commented. They cautioned, however, that rechallenge is not recommended for all patients, at least not for those with grade 2 or higher ILD/pneumonitis.
Overall, the authors concluded that the “benefit-risk of T-DXd treatment is positive,” but they warned that some patients may be at increased risk of developing ILD/pneumonitis
Baseline factors that increase the risk of developing an ILD/pneumonitis event include the following: being younger than 65 years, receiving a T-DXd dose of more than6.4 mg/kg, having a baseline oxygen saturation level of less than 95%, having moderate to severe renal impairment, and having lung comorbidities. In addition, patients who had initially been diagnosed with cancer more than 4 years before receiving the drug were at higher risk of developing ILD/pneumonitis.
“Using learnings from the early clinical trials experience, physician education and patient management protocols were revised and disseminated by the study sponsors [and] more recent trial data in earlier lines of therapy has demonstrated lower rates of ILD events, suggesting close monitoring and proactive management of ILD/pneumonitis is warranted for all patients,” Dr. Powell said in a statement.
The T-DXd clinical trials were sponsored by AstraZeneca and Daiichi Sankyo. Dr. Powell has received fees from Daiichi Sankyo, AstraZeneca, and Voluntis.
A version of this article first appeared on Medscape.com.
FROM ESMO OPEN
CT scan changes indicate increased mortality risk in ever-smokers
Longitudinal progression of parenchymal changes on CT images — also referred to as quantitative interstitial abnormalities (QIA) – is independently associated with decreased lung function and an increased all-cause mortality risk, an analysis of two cohorts of ever-smokers indicates. And among the main risk factors for QIA progression is smoking.
“These abnormalities have gone by a few different names but fundamentally, they are high density findings of chest CT that in some cases represent early or subtle evidence of pulmonary fibrosis,” Samuel Ash, MD, MPH, assistant professor of medicine, Brigham and Women’s Hospital, Boston, told this news organization.
So when I see someone with visual evidence of this type of change on their chest CT, I make sure to emphasize that while they don’t have interstitial lung disease [ILD] yet, these findings suggest they may be susceptible to lung injury from tobacco smoke and that if they don’t stop smoking now, they are at risk for a disease like interstitial pulmonary fibrosis [IPF] which is a highly morbid disease with a high mortality risk,” he added.
The study was published online in the journal CHEST.
Ever-smoking cohorts
Analysis of QIA progression on CT chest scans was carried out on participants from the Genetic Epidemiology of COPD (COPDGene) study as well as those from the Pittsburgh Lung Screening Study (PLuSS). COPDGene was a prospective cohort of over 10,300 ever-smokers with at least a 10–pack-year smoking history between the ages of 45 and 80. Participants underwent a series of tests including chest CT scans at baseline between 2006 and 2011 and again approximately 5 years later.
Patients with a postbronchodilator forced expiratory volume in 1 second (FEV1) of 80% or more of predicted and a FEV1-to-FVC (forced vital capacity) ratio of at least 0.7 were defined to have GOLD stage 0 disease while those with a postbronchodilator FEV1 of 80% or less than predicted and a FEV1-to-FVC ratio of at least 0.7 were defined to have preserved ratio impaired spirometry (PRISm) disease.
PLuSS involved 3,642 ever-smokers between the ages of 50 years and 79 years with at least a 12.5–pack-year history with no prior history of lung cancer. Participants again underwent a series of tests including a CT scan on visit 1 between 2002 and 2005 and then a second CT scan at a second visit almost 9 years later. “In the COPDGene cohort, 4,635 participants had complete clinical data, CT scans and spirometry from visits 1 and 2 for analysis,” the authors reported.
At visit 1 almost 48% of participants were current smokers and the mean pack-year history of the cohort was 41.9 years. The mean time between visits 1 and 2 was 5.6 years. Both the mean prebronchodilator FEV1 as well as the mean FVC decreased between visits 1 and 2. For example, the mean prebronchodilator FEV1 dropped from 2.2 liters to 2.0 liters between visits 1 and 2 while the mean prebronchodilator FVC decreased from 3.2 liters to 3.0 liters between the first and second visits.
In the PLuSS cohort, 1,307 participants had complete imaging and spirometry data available for visits 1 and 2 for analysis. The mean time between visits 1 and 2 was 8.6 years. Over 59% of the cohort were current smokers with a mean pack-year history of 65. Again, the mean prebronchodilator FEV1 and FVC both dropped between visit 1 and 2, as the authors note.
The mean prebronchodilator FEV1, for example, decreased from 2.5 liters to 2.1 liters between visits 1 and 2 while the mean prebronchodilator FVC dropped from 3.6 liters to 3.2 liters during the same interval. Looking at risk factors associated with QIA progression, investigators note that each additional year of baseline age was associated with a higher annual increase in QIA by 0.01% per year (95% confidence interval, 0.01%-0.02%; P < .001) in the COPDGene cohort and a 0.02% increase (95% CI, 0.01%-0.02%; P < .001) in the PLuSS cohort.
Female sex in turn was associated with a 0.07% per year (95% CI, 0.02%-0.12%; P = .003) higher increase in the QIA, compared with men in the COPDGene cohort and a 0.14% (95% CI, 0.02%-0.26%; P = .025) per year higher increase in the QIA in the PLuSS cohort. Current smoking status was only associated with a higher rate of QIA progression in the COPDGene cohort at a rate of 0.10% per year (95% CI, 0.06%-0.15%; P < .001).
Lastly, every copy of the minor allele of the MUIC5B promoter polymorphism was associated with a 0.12% per year (95% CI, 0.07%-0.16%; P < .0001) increase in QIA in the COPDGene cohort as well.
Smoking cessation
Smoking cessation is the obvious first step for patients with evidence of QIA progression but physicians can probably do more for these patients sooner, Dr. Ash said. “If we use heart disease as an analogy, we don’t want to start treating someone until they have a heart attack or are in heart failure, we start by checking their cholesterol and blood pressure and treating them with medications to prevent progression.”
Similarly, physicians need to start thinking about IPF and other lung diseases in the same way. For IPF, medications such as pirfenidone (Esbriet) and nintedanib (Ofev) do not reverse prior lung damage but they do slow disease progression and physicians need to initiate treatment before patients are short of breath, not after. Meantime, Dr. Ash advised physicians that, if they have a patient who is getting a CT scan for whatever reason, they should keep a close eye on whether or not patients have any of these interstitial changes and, if they do, then if the changes are getting worse.
“These patients are likely to be the ones who are going to develop IPF and who may benefit from ongoing imaging surveillance,” he said. And while clinicians may not yet be ready to use a quantitative tool at the bedside, “this tool – or one like it – is coming and we have to start thinking about how to incorporate these types of devices into our clinical practice.”
Temporal changes
Asked to comment on the findings, Surya Bhatt, MD, associate professor of medicine at the University of Alabama at Birmingham, said that the study advances the community’s understanding of the relationship between temporal changes in objectively measured interstitial lung abnormalities and several important clinical outcomes, including lung function decline and mortality. “Several risk factors for progression were also identified,” he noted.
“And these results make a case for initiating clinical trials to determine whether early treatment with existing antifibrotic medications in these high risk individuals can decrease the perpetuation of these permanent lung changes,” Dr. Bhatt said.
The COPDGene study was supported in part by contributions made by an industry advisory board. Dr. Ash was supported in part by Quantitative Imaging Solutions. Dr. Bhatt declared that he has receiving consulting fees or has service on advisory boards for Boehringer Ingelheim and Sanofi/Regeneron. He ha also received fee for CME from IntegrityCE.
Longitudinal progression of parenchymal changes on CT images — also referred to as quantitative interstitial abnormalities (QIA) – is independently associated with decreased lung function and an increased all-cause mortality risk, an analysis of two cohorts of ever-smokers indicates. And among the main risk factors for QIA progression is smoking.
“These abnormalities have gone by a few different names but fundamentally, they are high density findings of chest CT that in some cases represent early or subtle evidence of pulmonary fibrosis,” Samuel Ash, MD, MPH, assistant professor of medicine, Brigham and Women’s Hospital, Boston, told this news organization.
So when I see someone with visual evidence of this type of change on their chest CT, I make sure to emphasize that while they don’t have interstitial lung disease [ILD] yet, these findings suggest they may be susceptible to lung injury from tobacco smoke and that if they don’t stop smoking now, they are at risk for a disease like interstitial pulmonary fibrosis [IPF] which is a highly morbid disease with a high mortality risk,” he added.
The study was published online in the journal CHEST.
Ever-smoking cohorts
Analysis of QIA progression on CT chest scans was carried out on participants from the Genetic Epidemiology of COPD (COPDGene) study as well as those from the Pittsburgh Lung Screening Study (PLuSS). COPDGene was a prospective cohort of over 10,300 ever-smokers with at least a 10–pack-year smoking history between the ages of 45 and 80. Participants underwent a series of tests including chest CT scans at baseline between 2006 and 2011 and again approximately 5 years later.
Patients with a postbronchodilator forced expiratory volume in 1 second (FEV1) of 80% or more of predicted and a FEV1-to-FVC (forced vital capacity) ratio of at least 0.7 were defined to have GOLD stage 0 disease while those with a postbronchodilator FEV1 of 80% or less than predicted and a FEV1-to-FVC ratio of at least 0.7 were defined to have preserved ratio impaired spirometry (PRISm) disease.
PLuSS involved 3,642 ever-smokers between the ages of 50 years and 79 years with at least a 12.5–pack-year history with no prior history of lung cancer. Participants again underwent a series of tests including a CT scan on visit 1 between 2002 and 2005 and then a second CT scan at a second visit almost 9 years later. “In the COPDGene cohort, 4,635 participants had complete clinical data, CT scans and spirometry from visits 1 and 2 for analysis,” the authors reported.
At visit 1 almost 48% of participants were current smokers and the mean pack-year history of the cohort was 41.9 years. The mean time between visits 1 and 2 was 5.6 years. Both the mean prebronchodilator FEV1 as well as the mean FVC decreased between visits 1 and 2. For example, the mean prebronchodilator FEV1 dropped from 2.2 liters to 2.0 liters between visits 1 and 2 while the mean prebronchodilator FVC decreased from 3.2 liters to 3.0 liters between the first and second visits.
In the PLuSS cohort, 1,307 participants had complete imaging and spirometry data available for visits 1 and 2 for analysis. The mean time between visits 1 and 2 was 8.6 years. Over 59% of the cohort were current smokers with a mean pack-year history of 65. Again, the mean prebronchodilator FEV1 and FVC both dropped between visit 1 and 2, as the authors note.
The mean prebronchodilator FEV1, for example, decreased from 2.5 liters to 2.1 liters between visits 1 and 2 while the mean prebronchodilator FVC dropped from 3.6 liters to 3.2 liters during the same interval. Looking at risk factors associated with QIA progression, investigators note that each additional year of baseline age was associated with a higher annual increase in QIA by 0.01% per year (95% confidence interval, 0.01%-0.02%; P < .001) in the COPDGene cohort and a 0.02% increase (95% CI, 0.01%-0.02%; P < .001) in the PLuSS cohort.
Female sex in turn was associated with a 0.07% per year (95% CI, 0.02%-0.12%; P = .003) higher increase in the QIA, compared with men in the COPDGene cohort and a 0.14% (95% CI, 0.02%-0.26%; P = .025) per year higher increase in the QIA in the PLuSS cohort. Current smoking status was only associated with a higher rate of QIA progression in the COPDGene cohort at a rate of 0.10% per year (95% CI, 0.06%-0.15%; P < .001).
Lastly, every copy of the minor allele of the MUIC5B promoter polymorphism was associated with a 0.12% per year (95% CI, 0.07%-0.16%; P < .0001) increase in QIA in the COPDGene cohort as well.
Smoking cessation
Smoking cessation is the obvious first step for patients with evidence of QIA progression but physicians can probably do more for these patients sooner, Dr. Ash said. “If we use heart disease as an analogy, we don’t want to start treating someone until they have a heart attack or are in heart failure, we start by checking their cholesterol and blood pressure and treating them with medications to prevent progression.”
Similarly, physicians need to start thinking about IPF and other lung diseases in the same way. For IPF, medications such as pirfenidone (Esbriet) and nintedanib (Ofev) do not reverse prior lung damage but they do slow disease progression and physicians need to initiate treatment before patients are short of breath, not after. Meantime, Dr. Ash advised physicians that, if they have a patient who is getting a CT scan for whatever reason, they should keep a close eye on whether or not patients have any of these interstitial changes and, if they do, then if the changes are getting worse.
“These patients are likely to be the ones who are going to develop IPF and who may benefit from ongoing imaging surveillance,” he said. And while clinicians may not yet be ready to use a quantitative tool at the bedside, “this tool – or one like it – is coming and we have to start thinking about how to incorporate these types of devices into our clinical practice.”
Temporal changes
Asked to comment on the findings, Surya Bhatt, MD, associate professor of medicine at the University of Alabama at Birmingham, said that the study advances the community’s understanding of the relationship between temporal changes in objectively measured interstitial lung abnormalities and several important clinical outcomes, including lung function decline and mortality. “Several risk factors for progression were also identified,” he noted.
“And these results make a case for initiating clinical trials to determine whether early treatment with existing antifibrotic medications in these high risk individuals can decrease the perpetuation of these permanent lung changes,” Dr. Bhatt said.
The COPDGene study was supported in part by contributions made by an industry advisory board. Dr. Ash was supported in part by Quantitative Imaging Solutions. Dr. Bhatt declared that he has receiving consulting fees or has service on advisory boards for Boehringer Ingelheim and Sanofi/Regeneron. He ha also received fee for CME from IntegrityCE.
Longitudinal progression of parenchymal changes on CT images — also referred to as quantitative interstitial abnormalities (QIA) – is independently associated with decreased lung function and an increased all-cause mortality risk, an analysis of two cohorts of ever-smokers indicates. And among the main risk factors for QIA progression is smoking.
“These abnormalities have gone by a few different names but fundamentally, they are high density findings of chest CT that in some cases represent early or subtle evidence of pulmonary fibrosis,” Samuel Ash, MD, MPH, assistant professor of medicine, Brigham and Women’s Hospital, Boston, told this news organization.
So when I see someone with visual evidence of this type of change on their chest CT, I make sure to emphasize that while they don’t have interstitial lung disease [ILD] yet, these findings suggest they may be susceptible to lung injury from tobacco smoke and that if they don’t stop smoking now, they are at risk for a disease like interstitial pulmonary fibrosis [IPF] which is a highly morbid disease with a high mortality risk,” he added.
The study was published online in the journal CHEST.
Ever-smoking cohorts
Analysis of QIA progression on CT chest scans was carried out on participants from the Genetic Epidemiology of COPD (COPDGene) study as well as those from the Pittsburgh Lung Screening Study (PLuSS). COPDGene was a prospective cohort of over 10,300 ever-smokers with at least a 10–pack-year smoking history between the ages of 45 and 80. Participants underwent a series of tests including chest CT scans at baseline between 2006 and 2011 and again approximately 5 years later.
Patients with a postbronchodilator forced expiratory volume in 1 second (FEV1) of 80% or more of predicted and a FEV1-to-FVC (forced vital capacity) ratio of at least 0.7 were defined to have GOLD stage 0 disease while those with a postbronchodilator FEV1 of 80% or less than predicted and a FEV1-to-FVC ratio of at least 0.7 were defined to have preserved ratio impaired spirometry (PRISm) disease.
PLuSS involved 3,642 ever-smokers between the ages of 50 years and 79 years with at least a 12.5–pack-year history with no prior history of lung cancer. Participants again underwent a series of tests including a CT scan on visit 1 between 2002 and 2005 and then a second CT scan at a second visit almost 9 years later. “In the COPDGene cohort, 4,635 participants had complete clinical data, CT scans and spirometry from visits 1 and 2 for analysis,” the authors reported.
At visit 1 almost 48% of participants were current smokers and the mean pack-year history of the cohort was 41.9 years. The mean time between visits 1 and 2 was 5.6 years. Both the mean prebronchodilator FEV1 as well as the mean FVC decreased between visits 1 and 2. For example, the mean prebronchodilator FEV1 dropped from 2.2 liters to 2.0 liters between visits 1 and 2 while the mean prebronchodilator FVC decreased from 3.2 liters to 3.0 liters between the first and second visits.
In the PLuSS cohort, 1,307 participants had complete imaging and spirometry data available for visits 1 and 2 for analysis. The mean time between visits 1 and 2 was 8.6 years. Over 59% of the cohort were current smokers with a mean pack-year history of 65. Again, the mean prebronchodilator FEV1 and FVC both dropped between visit 1 and 2, as the authors note.
The mean prebronchodilator FEV1, for example, decreased from 2.5 liters to 2.1 liters between visits 1 and 2 while the mean prebronchodilator FVC dropped from 3.6 liters to 3.2 liters during the same interval. Looking at risk factors associated with QIA progression, investigators note that each additional year of baseline age was associated with a higher annual increase in QIA by 0.01% per year (95% confidence interval, 0.01%-0.02%; P < .001) in the COPDGene cohort and a 0.02% increase (95% CI, 0.01%-0.02%; P < .001) in the PLuSS cohort.
Female sex in turn was associated with a 0.07% per year (95% CI, 0.02%-0.12%; P = .003) higher increase in the QIA, compared with men in the COPDGene cohort and a 0.14% (95% CI, 0.02%-0.26%; P = .025) per year higher increase in the QIA in the PLuSS cohort. Current smoking status was only associated with a higher rate of QIA progression in the COPDGene cohort at a rate of 0.10% per year (95% CI, 0.06%-0.15%; P < .001).
Lastly, every copy of the minor allele of the MUIC5B promoter polymorphism was associated with a 0.12% per year (95% CI, 0.07%-0.16%; P < .0001) increase in QIA in the COPDGene cohort as well.
Smoking cessation
Smoking cessation is the obvious first step for patients with evidence of QIA progression but physicians can probably do more for these patients sooner, Dr. Ash said. “If we use heart disease as an analogy, we don’t want to start treating someone until they have a heart attack or are in heart failure, we start by checking their cholesterol and blood pressure and treating them with medications to prevent progression.”
Similarly, physicians need to start thinking about IPF and other lung diseases in the same way. For IPF, medications such as pirfenidone (Esbriet) and nintedanib (Ofev) do not reverse prior lung damage but they do slow disease progression and physicians need to initiate treatment before patients are short of breath, not after. Meantime, Dr. Ash advised physicians that, if they have a patient who is getting a CT scan for whatever reason, they should keep a close eye on whether or not patients have any of these interstitial changes and, if they do, then if the changes are getting worse.
“These patients are likely to be the ones who are going to develop IPF and who may benefit from ongoing imaging surveillance,” he said. And while clinicians may not yet be ready to use a quantitative tool at the bedside, “this tool – or one like it – is coming and we have to start thinking about how to incorporate these types of devices into our clinical practice.”
Temporal changes
Asked to comment on the findings, Surya Bhatt, MD, associate professor of medicine at the University of Alabama at Birmingham, said that the study advances the community’s understanding of the relationship between temporal changes in objectively measured interstitial lung abnormalities and several important clinical outcomes, including lung function decline and mortality. “Several risk factors for progression were also identified,” he noted.
“And these results make a case for initiating clinical trials to determine whether early treatment with existing antifibrotic medications in these high risk individuals can decrease the perpetuation of these permanent lung changes,” Dr. Bhatt said.
The COPDGene study was supported in part by contributions made by an industry advisory board. Dr. Ash was supported in part by Quantitative Imaging Solutions. Dr. Bhatt declared that he has receiving consulting fees or has service on advisory boards for Boehringer Ingelheim and Sanofi/Regeneron. He ha also received fee for CME from IntegrityCE.
FROM CHEST
Genetic counseling for cancer often costs patients nothing
But even among those who do, the cost is $16 or less, a cohort study shows.
“The findings highlight the relatively low financial costs of genetic counseling, a form of care with potentially substantial implications for cancer treatment,” lead author Mya Roberson, PhD, Vanderbilt University, Nashville, Tenn., and colleagues explained.
The study was published online in JAMA Health Forum.
Genetic counseling is an important feature of cancer care that can affect treatment decisions and surveillance. But coverage of genetic counseling services varies across insurance types.
To understand the costs to patients, the investigators used data from IBM Watson Health MarketScan to create a cohort of privately insured patients with breast, prostate, endometrial, ovarian, colorectal, and pancreatic cancer who had at least one genetic counseling session from 2013 to the end of 2019.
Dr. Roberson and colleagues then calculated out-of-pocket costs – including coinsurance, copayments, and deductibles – and total costs paid on claims for genetic counseling encounters. The cohort included 16,791 patients, the majority of whom had breast cancer.
Although the median insurance payment for genetic counseling encounters was $118 ($58-$211), most patients paid nothing out of pocket for these services. Among the 31% of patients with an out-of-pocket expense, the cost was $16 or less.
Compared with breast cancer patients, men with prostate cancer were 28% more likely to have out-of-pocket costs for genetic counseling, which may “reflect a lack of awareness about the medical necessity of genetic counseling,” the authors suggested.
Overall, the study highlights the value of genetic counseling in cancer care.
“Cancer genetic counseling not only promotes informed decision-making about genetic testing and cancer treatment in the era of precision medicine, but it also is a form of low-cost, high-value care,” the authors wrote.
The study was funded by a grant from the National Cancer Institute. Dr. Roberson disclosed no relevant financial relationships.
A version of this article first appeared on Medscape.com.
But even among those who do, the cost is $16 or less, a cohort study shows.
“The findings highlight the relatively low financial costs of genetic counseling, a form of care with potentially substantial implications for cancer treatment,” lead author Mya Roberson, PhD, Vanderbilt University, Nashville, Tenn., and colleagues explained.
The study was published online in JAMA Health Forum.
Genetic counseling is an important feature of cancer care that can affect treatment decisions and surveillance. But coverage of genetic counseling services varies across insurance types.
To understand the costs to patients, the investigators used data from IBM Watson Health MarketScan to create a cohort of privately insured patients with breast, prostate, endometrial, ovarian, colorectal, and pancreatic cancer who had at least one genetic counseling session from 2013 to the end of 2019.
Dr. Roberson and colleagues then calculated out-of-pocket costs – including coinsurance, copayments, and deductibles – and total costs paid on claims for genetic counseling encounters. The cohort included 16,791 patients, the majority of whom had breast cancer.
Although the median insurance payment for genetic counseling encounters was $118 ($58-$211), most patients paid nothing out of pocket for these services. Among the 31% of patients with an out-of-pocket expense, the cost was $16 or less.
Compared with breast cancer patients, men with prostate cancer were 28% more likely to have out-of-pocket costs for genetic counseling, which may “reflect a lack of awareness about the medical necessity of genetic counseling,” the authors suggested.
Overall, the study highlights the value of genetic counseling in cancer care.
“Cancer genetic counseling not only promotes informed decision-making about genetic testing and cancer treatment in the era of precision medicine, but it also is a form of low-cost, high-value care,” the authors wrote.
The study was funded by a grant from the National Cancer Institute. Dr. Roberson disclosed no relevant financial relationships.
A version of this article first appeared on Medscape.com.
But even among those who do, the cost is $16 or less, a cohort study shows.
“The findings highlight the relatively low financial costs of genetic counseling, a form of care with potentially substantial implications for cancer treatment,” lead author Mya Roberson, PhD, Vanderbilt University, Nashville, Tenn., and colleagues explained.
The study was published online in JAMA Health Forum.
Genetic counseling is an important feature of cancer care that can affect treatment decisions and surveillance. But coverage of genetic counseling services varies across insurance types.
To understand the costs to patients, the investigators used data from IBM Watson Health MarketScan to create a cohort of privately insured patients with breast, prostate, endometrial, ovarian, colorectal, and pancreatic cancer who had at least one genetic counseling session from 2013 to the end of 2019.
Dr. Roberson and colleagues then calculated out-of-pocket costs – including coinsurance, copayments, and deductibles – and total costs paid on claims for genetic counseling encounters. The cohort included 16,791 patients, the majority of whom had breast cancer.
Although the median insurance payment for genetic counseling encounters was $118 ($58-$211), most patients paid nothing out of pocket for these services. Among the 31% of patients with an out-of-pocket expense, the cost was $16 or less.
Compared with breast cancer patients, men with prostate cancer were 28% more likely to have out-of-pocket costs for genetic counseling, which may “reflect a lack of awareness about the medical necessity of genetic counseling,” the authors suggested.
Overall, the study highlights the value of genetic counseling in cancer care.
“Cancer genetic counseling not only promotes informed decision-making about genetic testing and cancer treatment in the era of precision medicine, but it also is a form of low-cost, high-value care,” the authors wrote.
The study was funded by a grant from the National Cancer Institute. Dr. Roberson disclosed no relevant financial relationships.
A version of this article first appeared on Medscape.com.
FROM JAMA HEALTH FORUM
Updates on treatment/prevention of VTE in cancer patients
Updated clinical practice guidelines for the treatment and prevention of venous thromboembolism for patients with cancer, including those with cancer and COVID-19, have been released by the International Initiative on Thrombosis and Cancer (ITAC), an academic working group of VTE experts.
“Because patients with cancer have a baseline increased risk of VTE, compared with patients without cancer, the combination of both COVID-19 and cancer – and its effect on VTE risk and treatment – is of concern,” said the authors, led by Dominique Farge, MD, PhD, Nord Universite de Paris.
they added.
The new guidelines were published online in The Lancet Oncology.
“Cancer-associated VTE remains an important clinical problem, associated with increased morbidity and mortality,” Dr. Farge and colleagues observed.
“The ITAC guidelines’ companion free web-based mobile application will assist the practicing clinician with decision making at various levels to provide optimal care of patients with cancer to treat and prevent VTE,” they emphasized. More information is available at itaccme.com.
Cancer patients with COVID
The new section of the guidelines notes that the treatment and prevention of VTE for cancer patients infected with SARS-CoV-2 remain the same as for patients without COVID.
Whether or not cancer patients with COVID-19 are hospitalized, have been discharged, or are ambulatory, they should be assessed for the risk of VTE, as should any other patient. For cancer patients with COVID-19 who are hospitalized, pharmacologic prophylaxis should be given at the same dose and anticoagulant type as for hospitalized cancer patients who do not have COVID-19.
Following discharge, VTE prophylaxis is not advised for cancer patients infected with SARS-CoV-2, and routine primary pharmacologic prophylaxis of VTE for ambulatory patients with COVID-19 is also not recommended, the authors noted.
Initial treatment of established VTE
Initial treatment of established VTE for up to 10 days of anticoagulation should include low-molecular-weight heparin (LMWH) when creatinine clearance is at least 30 mL/min.
“A regimen of LMWH, taken once per day, is recommended unless a twice-per-day regimen is required because of patients’ characteristics,” the authors noted. These characteristics include a high risk of bleeding, moderate renal failure, and the need for technical intervention, including surgery.
If a twice-a-day regimen is required, only enoxaparin at a dose of 1 mg/kg twice daily can be used, the authors cautioned.
For patients with a low risk of gastrointestinal or genitourinary bleeding, rivaroxaban (Xarelto) or apixaban (Eliquis) can be given in the first 10 days, as well as edoxaban (Lixiana). The latter should be started after at least 5 days of parenteral anticoagulation, provided creatinine clearance is at least 30 mL/min.
“Unfractionated heparin as well as fondaparinux (GlaxoSmithKline) can be also used for the initial treatment of established VTE when LMWH or direct oral anticoagulants are contraindicated,” Dr. Farge and colleagues wrote.
Thrombolysis can be considered on a case-by-case basis, although physicians must pay attention to specific contraindications, especially bleeding risk.
“In the initial treatment of VTE, inferior vena cava filters might be considered when anticoagulant treatment is contraindicated or, in the case of pulmonary embolism, when recurrence occurs under optimal anticoagulation,” the authors noted.
Maintenance VTE treatment
For maintenance therapy, which the authors define as early maintenance for up to 6 months and long-term maintenance beyond 6 months, they point out that LMWHs are preferred over vitamin K antagonists for the treatment of VTE when the creatinine clearance is again at least 30 mL/min.
Any of the direct oral anticoagulants (DOAs) – edoxaban, rivaroxaban, or apixaban – is also recommended for the same patients, provided there is no risk of inducing a strong drug-drug interaction or GI absorption is impaired.
However, the DOAs should be used with caution for patients with GI malignancies, especially upper GI cancers, because data show there is an increased risk of GI bleeding with both edoxaban and rivaroxaban.
“LMWH or direct oral anticoagulants should be used for a minimum of 6 months to treat established VTE in patients with cancer,” the authors wrote.
“After 6 months, termination or continuation of anticoagulation (LMWH, direct oral anticoagulants, or vitamin K antagonists) should be based on individual evaluation of the benefit-risk ratio,” they added.
Treatment of VTE recurrence
The guideline authors explain that three options can be considered in the event of VTE recurrence. These include an increase in the LMWH dose by 20%-25%, or a switch to a DOA, or, if patients are taking a DOA, a switch to an LMWH. If the patient is taking a vitamin K antagonist, it can be switched to either an LMWH or a DOA.
For treatment of catheter-related thrombosis, anticoagulant treatment is recommended for a minimum of 3 months and as long as the central venous catheter is in place. In this setting, the LMWHs are recommended.
The central venous catheter can be kept in place if it is functional, well positioned, and is not infected, provided there is good resolution of symptoms under close surveillance while anticoagulants are being administered.
In surgically treated patients, the LMWH, given once a day, to patients with a serum creatinine concentration of at least 30 mL/min can be used to prevent VTE. Alternatively, VTE can be prevented by the use low-dose unfractionated heparin, given three times a day.
“Pharmacological prophylaxis should be started 2-12 h preoperatively and continued for at least 7–10 days,” Dr. Farge and colleagues advised. In this setting, there is insufficient evidence to support the use of fondaparinux or a DOA as an alternative to an LMWH for the prophylaxis of postoperative VTE. “Use of the highest prophylactic dose of LMWH to prevent postoperative VTE in patients with cancer is recommended,” the authors advised.
Furthermore, extended prophylaxis of at least 4 weeks with LMWH is advised to prevent postoperative VTE after major abdominal or pelvic surgery. Mechanical methods are not recommended except when pharmacologic methods are contraindicated. Inferior vena cava filters are also not recommended for routine prophylaxis.
Patients with reduced mobility
For medically treated hospitalized patients with cancer whose mobility is reduced, the authors recommend prophylaxis with either an LMWH or fondaparinux, provided their creatinine clearance is at least 30 mL/min. These patients can also be treated with unfractionated heparin, they add.
In contrast, DOAs are not recommended – at least not routinely – in this setting, the authors cautioned. Primary pharmacologic prophylaxis of VTE with either LMWH or DOAs – either rivaroxaban or apixaban – is indicated in ambulatory patients with locally advanced or metastatic pancreatic cancer who are receiving systemic anticancer therapy, provided they are at low risk of bleeding.
However, primary pharmacologic prophylaxis with LMWH is not recommended outside of a clinical trial for patients with locally advanced or metastatic lung cancer who are undergoing systemic anticancer therapy, even for patients who are at low risk of bleeding.
For ambulatory patients who are receiving systemic anticancer therapy and who are at intermediate risk of VTE, primary prophylaxis with rivaroxaban or apixaban is recommended for those with myeloma who are receiving immunomodulatory therapy plus steroids or other systemic therapies.
In this setting, oral anticoagulants should consist of a vitamin K antagonist, given at low or therapeutic doses, or apixaban, given at prophylactic doses. Alternatively, LMWH, given at prophylactic doses, or low-dose aspirin, given at a dose of 100 mg/day, can be used.
Catheter-related thrombosis
Use of anticoagulation for routine prophylaxis of catheter-related thrombosis is not recommended. Catheters should be inserted on the right side in the jugular vein, and the distal extremity of the central catheter should be located at the junction of the superior vena cava and the right atrium. “In patients requiring central venous catheters, we suggest the use of implanted ports over peripheral inserted central catheter lines,” the authors noted.
The authors described a number of unique situations regarding the treatment of VTE. These situations include patients with a brain tumor, for whom treatment of established VTE should favor either LMWH or a DOA. The authors also recommended the use of LMWH or unfractionated heparin, started postoperatively, for the prevention of VTE for patients undergoing neurosurgery.
In contrast, pharmacologic prophylaxis of VTE in medically treated patients with a brain tumor who are not undergoing neurosurgery is not recommended. “In the presence of severe renal failure...we suggest using unfractionated heparin followed by early vitamin K antagonists (possibly from day 1) or LMWH adjusted to anti-Xa concentration of the treatment of established VTE,” Dr. Farge and colleagues wrote.
Anticoagulant treatment is also recommended for a minimum of 3 months for children with symptomatic catheter-related thrombosis and as long as the central venous catheter is in place. For children with acute lymphoblastic leukemia who are undergoing induction chemotherapy, LMWH is also recommended as thromboprophylaxis.
For children who require a central venous catheter, the authors suggested that physicians use implanted ports over peripherally inserted central lines.
A version of this article first appeared on Medscape.com.
Updated clinical practice guidelines for the treatment and prevention of venous thromboembolism for patients with cancer, including those with cancer and COVID-19, have been released by the International Initiative on Thrombosis and Cancer (ITAC), an academic working group of VTE experts.
“Because patients with cancer have a baseline increased risk of VTE, compared with patients without cancer, the combination of both COVID-19 and cancer – and its effect on VTE risk and treatment – is of concern,” said the authors, led by Dominique Farge, MD, PhD, Nord Universite de Paris.
they added.
The new guidelines were published online in The Lancet Oncology.
“Cancer-associated VTE remains an important clinical problem, associated with increased morbidity and mortality,” Dr. Farge and colleagues observed.
“The ITAC guidelines’ companion free web-based mobile application will assist the practicing clinician with decision making at various levels to provide optimal care of patients with cancer to treat and prevent VTE,” they emphasized. More information is available at itaccme.com.
Cancer patients with COVID
The new section of the guidelines notes that the treatment and prevention of VTE for cancer patients infected with SARS-CoV-2 remain the same as for patients without COVID.
Whether or not cancer patients with COVID-19 are hospitalized, have been discharged, or are ambulatory, they should be assessed for the risk of VTE, as should any other patient. For cancer patients with COVID-19 who are hospitalized, pharmacologic prophylaxis should be given at the same dose and anticoagulant type as for hospitalized cancer patients who do not have COVID-19.
Following discharge, VTE prophylaxis is not advised for cancer patients infected with SARS-CoV-2, and routine primary pharmacologic prophylaxis of VTE for ambulatory patients with COVID-19 is also not recommended, the authors noted.
Initial treatment of established VTE
Initial treatment of established VTE for up to 10 days of anticoagulation should include low-molecular-weight heparin (LMWH) when creatinine clearance is at least 30 mL/min.
“A regimen of LMWH, taken once per day, is recommended unless a twice-per-day regimen is required because of patients’ characteristics,” the authors noted. These characteristics include a high risk of bleeding, moderate renal failure, and the need for technical intervention, including surgery.
If a twice-a-day regimen is required, only enoxaparin at a dose of 1 mg/kg twice daily can be used, the authors cautioned.
For patients with a low risk of gastrointestinal or genitourinary bleeding, rivaroxaban (Xarelto) or apixaban (Eliquis) can be given in the first 10 days, as well as edoxaban (Lixiana). The latter should be started after at least 5 days of parenteral anticoagulation, provided creatinine clearance is at least 30 mL/min.
“Unfractionated heparin as well as fondaparinux (GlaxoSmithKline) can be also used for the initial treatment of established VTE when LMWH or direct oral anticoagulants are contraindicated,” Dr. Farge and colleagues wrote.
Thrombolysis can be considered on a case-by-case basis, although physicians must pay attention to specific contraindications, especially bleeding risk.
“In the initial treatment of VTE, inferior vena cava filters might be considered when anticoagulant treatment is contraindicated or, in the case of pulmonary embolism, when recurrence occurs under optimal anticoagulation,” the authors noted.
Maintenance VTE treatment
For maintenance therapy, which the authors define as early maintenance for up to 6 months and long-term maintenance beyond 6 months, they point out that LMWHs are preferred over vitamin K antagonists for the treatment of VTE when the creatinine clearance is again at least 30 mL/min.
Any of the direct oral anticoagulants (DOAs) – edoxaban, rivaroxaban, or apixaban – is also recommended for the same patients, provided there is no risk of inducing a strong drug-drug interaction or GI absorption is impaired.
However, the DOAs should be used with caution for patients with GI malignancies, especially upper GI cancers, because data show there is an increased risk of GI bleeding with both edoxaban and rivaroxaban.
“LMWH or direct oral anticoagulants should be used for a minimum of 6 months to treat established VTE in patients with cancer,” the authors wrote.
“After 6 months, termination or continuation of anticoagulation (LMWH, direct oral anticoagulants, or vitamin K antagonists) should be based on individual evaluation of the benefit-risk ratio,” they added.
Treatment of VTE recurrence
The guideline authors explain that three options can be considered in the event of VTE recurrence. These include an increase in the LMWH dose by 20%-25%, or a switch to a DOA, or, if patients are taking a DOA, a switch to an LMWH. If the patient is taking a vitamin K antagonist, it can be switched to either an LMWH or a DOA.
For treatment of catheter-related thrombosis, anticoagulant treatment is recommended for a minimum of 3 months and as long as the central venous catheter is in place. In this setting, the LMWHs are recommended.
The central venous catheter can be kept in place if it is functional, well positioned, and is not infected, provided there is good resolution of symptoms under close surveillance while anticoagulants are being administered.
In surgically treated patients, the LMWH, given once a day, to patients with a serum creatinine concentration of at least 30 mL/min can be used to prevent VTE. Alternatively, VTE can be prevented by the use low-dose unfractionated heparin, given three times a day.
“Pharmacological prophylaxis should be started 2-12 h preoperatively and continued for at least 7–10 days,” Dr. Farge and colleagues advised. In this setting, there is insufficient evidence to support the use of fondaparinux or a DOA as an alternative to an LMWH for the prophylaxis of postoperative VTE. “Use of the highest prophylactic dose of LMWH to prevent postoperative VTE in patients with cancer is recommended,” the authors advised.
Furthermore, extended prophylaxis of at least 4 weeks with LMWH is advised to prevent postoperative VTE after major abdominal or pelvic surgery. Mechanical methods are not recommended except when pharmacologic methods are contraindicated. Inferior vena cava filters are also not recommended for routine prophylaxis.
Patients with reduced mobility
For medically treated hospitalized patients with cancer whose mobility is reduced, the authors recommend prophylaxis with either an LMWH or fondaparinux, provided their creatinine clearance is at least 30 mL/min. These patients can also be treated with unfractionated heparin, they add.
In contrast, DOAs are not recommended – at least not routinely – in this setting, the authors cautioned. Primary pharmacologic prophylaxis of VTE with either LMWH or DOAs – either rivaroxaban or apixaban – is indicated in ambulatory patients with locally advanced or metastatic pancreatic cancer who are receiving systemic anticancer therapy, provided they are at low risk of bleeding.
However, primary pharmacologic prophylaxis with LMWH is not recommended outside of a clinical trial for patients with locally advanced or metastatic lung cancer who are undergoing systemic anticancer therapy, even for patients who are at low risk of bleeding.
For ambulatory patients who are receiving systemic anticancer therapy and who are at intermediate risk of VTE, primary prophylaxis with rivaroxaban or apixaban is recommended for those with myeloma who are receiving immunomodulatory therapy plus steroids or other systemic therapies.
In this setting, oral anticoagulants should consist of a vitamin K antagonist, given at low or therapeutic doses, or apixaban, given at prophylactic doses. Alternatively, LMWH, given at prophylactic doses, or low-dose aspirin, given at a dose of 100 mg/day, can be used.
Catheter-related thrombosis
Use of anticoagulation for routine prophylaxis of catheter-related thrombosis is not recommended. Catheters should be inserted on the right side in the jugular vein, and the distal extremity of the central catheter should be located at the junction of the superior vena cava and the right atrium. “In patients requiring central venous catheters, we suggest the use of implanted ports over peripheral inserted central catheter lines,” the authors noted.
The authors described a number of unique situations regarding the treatment of VTE. These situations include patients with a brain tumor, for whom treatment of established VTE should favor either LMWH or a DOA. The authors also recommended the use of LMWH or unfractionated heparin, started postoperatively, for the prevention of VTE for patients undergoing neurosurgery.
In contrast, pharmacologic prophylaxis of VTE in medically treated patients with a brain tumor who are not undergoing neurosurgery is not recommended. “In the presence of severe renal failure...we suggest using unfractionated heparin followed by early vitamin K antagonists (possibly from day 1) or LMWH adjusted to anti-Xa concentration of the treatment of established VTE,” Dr. Farge and colleagues wrote.
Anticoagulant treatment is also recommended for a minimum of 3 months for children with symptomatic catheter-related thrombosis and as long as the central venous catheter is in place. For children with acute lymphoblastic leukemia who are undergoing induction chemotherapy, LMWH is also recommended as thromboprophylaxis.
For children who require a central venous catheter, the authors suggested that physicians use implanted ports over peripherally inserted central lines.
A version of this article first appeared on Medscape.com.
Updated clinical practice guidelines for the treatment and prevention of venous thromboembolism for patients with cancer, including those with cancer and COVID-19, have been released by the International Initiative on Thrombosis and Cancer (ITAC), an academic working group of VTE experts.
“Because patients with cancer have a baseline increased risk of VTE, compared with patients without cancer, the combination of both COVID-19 and cancer – and its effect on VTE risk and treatment – is of concern,” said the authors, led by Dominique Farge, MD, PhD, Nord Universite de Paris.
they added.
The new guidelines were published online in The Lancet Oncology.
“Cancer-associated VTE remains an important clinical problem, associated with increased morbidity and mortality,” Dr. Farge and colleagues observed.
“The ITAC guidelines’ companion free web-based mobile application will assist the practicing clinician with decision making at various levels to provide optimal care of patients with cancer to treat and prevent VTE,” they emphasized. More information is available at itaccme.com.
Cancer patients with COVID
The new section of the guidelines notes that the treatment and prevention of VTE for cancer patients infected with SARS-CoV-2 remain the same as for patients without COVID.
Whether or not cancer patients with COVID-19 are hospitalized, have been discharged, or are ambulatory, they should be assessed for the risk of VTE, as should any other patient. For cancer patients with COVID-19 who are hospitalized, pharmacologic prophylaxis should be given at the same dose and anticoagulant type as for hospitalized cancer patients who do not have COVID-19.
Following discharge, VTE prophylaxis is not advised for cancer patients infected with SARS-CoV-2, and routine primary pharmacologic prophylaxis of VTE for ambulatory patients with COVID-19 is also not recommended, the authors noted.
Initial treatment of established VTE
Initial treatment of established VTE for up to 10 days of anticoagulation should include low-molecular-weight heparin (LMWH) when creatinine clearance is at least 30 mL/min.
“A regimen of LMWH, taken once per day, is recommended unless a twice-per-day regimen is required because of patients’ characteristics,” the authors noted. These characteristics include a high risk of bleeding, moderate renal failure, and the need for technical intervention, including surgery.
If a twice-a-day regimen is required, only enoxaparin at a dose of 1 mg/kg twice daily can be used, the authors cautioned.
For patients with a low risk of gastrointestinal or genitourinary bleeding, rivaroxaban (Xarelto) or apixaban (Eliquis) can be given in the first 10 days, as well as edoxaban (Lixiana). The latter should be started after at least 5 days of parenteral anticoagulation, provided creatinine clearance is at least 30 mL/min.
“Unfractionated heparin as well as fondaparinux (GlaxoSmithKline) can be also used for the initial treatment of established VTE when LMWH or direct oral anticoagulants are contraindicated,” Dr. Farge and colleagues wrote.
Thrombolysis can be considered on a case-by-case basis, although physicians must pay attention to specific contraindications, especially bleeding risk.
“In the initial treatment of VTE, inferior vena cava filters might be considered when anticoagulant treatment is contraindicated or, in the case of pulmonary embolism, when recurrence occurs under optimal anticoagulation,” the authors noted.
Maintenance VTE treatment
For maintenance therapy, which the authors define as early maintenance for up to 6 months and long-term maintenance beyond 6 months, they point out that LMWHs are preferred over vitamin K antagonists for the treatment of VTE when the creatinine clearance is again at least 30 mL/min.
Any of the direct oral anticoagulants (DOAs) – edoxaban, rivaroxaban, or apixaban – is also recommended for the same patients, provided there is no risk of inducing a strong drug-drug interaction or GI absorption is impaired.
However, the DOAs should be used with caution for patients with GI malignancies, especially upper GI cancers, because data show there is an increased risk of GI bleeding with both edoxaban and rivaroxaban.
“LMWH or direct oral anticoagulants should be used for a minimum of 6 months to treat established VTE in patients with cancer,” the authors wrote.
“After 6 months, termination or continuation of anticoagulation (LMWH, direct oral anticoagulants, or vitamin K antagonists) should be based on individual evaluation of the benefit-risk ratio,” they added.
Treatment of VTE recurrence
The guideline authors explain that three options can be considered in the event of VTE recurrence. These include an increase in the LMWH dose by 20%-25%, or a switch to a DOA, or, if patients are taking a DOA, a switch to an LMWH. If the patient is taking a vitamin K antagonist, it can be switched to either an LMWH or a DOA.
For treatment of catheter-related thrombosis, anticoagulant treatment is recommended for a minimum of 3 months and as long as the central venous catheter is in place. In this setting, the LMWHs are recommended.
The central venous catheter can be kept in place if it is functional, well positioned, and is not infected, provided there is good resolution of symptoms under close surveillance while anticoagulants are being administered.
In surgically treated patients, the LMWH, given once a day, to patients with a serum creatinine concentration of at least 30 mL/min can be used to prevent VTE. Alternatively, VTE can be prevented by the use low-dose unfractionated heparin, given three times a day.
“Pharmacological prophylaxis should be started 2-12 h preoperatively and continued for at least 7–10 days,” Dr. Farge and colleagues advised. In this setting, there is insufficient evidence to support the use of fondaparinux or a DOA as an alternative to an LMWH for the prophylaxis of postoperative VTE. “Use of the highest prophylactic dose of LMWH to prevent postoperative VTE in patients with cancer is recommended,” the authors advised.
Furthermore, extended prophylaxis of at least 4 weeks with LMWH is advised to prevent postoperative VTE after major abdominal or pelvic surgery. Mechanical methods are not recommended except when pharmacologic methods are contraindicated. Inferior vena cava filters are also not recommended for routine prophylaxis.
Patients with reduced mobility
For medically treated hospitalized patients with cancer whose mobility is reduced, the authors recommend prophylaxis with either an LMWH or fondaparinux, provided their creatinine clearance is at least 30 mL/min. These patients can also be treated with unfractionated heparin, they add.
In contrast, DOAs are not recommended – at least not routinely – in this setting, the authors cautioned. Primary pharmacologic prophylaxis of VTE with either LMWH or DOAs – either rivaroxaban or apixaban – is indicated in ambulatory patients with locally advanced or metastatic pancreatic cancer who are receiving systemic anticancer therapy, provided they are at low risk of bleeding.
However, primary pharmacologic prophylaxis with LMWH is not recommended outside of a clinical trial for patients with locally advanced or metastatic lung cancer who are undergoing systemic anticancer therapy, even for patients who are at low risk of bleeding.
For ambulatory patients who are receiving systemic anticancer therapy and who are at intermediate risk of VTE, primary prophylaxis with rivaroxaban or apixaban is recommended for those with myeloma who are receiving immunomodulatory therapy plus steroids or other systemic therapies.
In this setting, oral anticoagulants should consist of a vitamin K antagonist, given at low or therapeutic doses, or apixaban, given at prophylactic doses. Alternatively, LMWH, given at prophylactic doses, or low-dose aspirin, given at a dose of 100 mg/day, can be used.
Catheter-related thrombosis
Use of anticoagulation for routine prophylaxis of catheter-related thrombosis is not recommended. Catheters should be inserted on the right side in the jugular vein, and the distal extremity of the central catheter should be located at the junction of the superior vena cava and the right atrium. “In patients requiring central venous catheters, we suggest the use of implanted ports over peripheral inserted central catheter lines,” the authors noted.
The authors described a number of unique situations regarding the treatment of VTE. These situations include patients with a brain tumor, for whom treatment of established VTE should favor either LMWH or a DOA. The authors also recommended the use of LMWH or unfractionated heparin, started postoperatively, for the prevention of VTE for patients undergoing neurosurgery.
In contrast, pharmacologic prophylaxis of VTE in medically treated patients with a brain tumor who are not undergoing neurosurgery is not recommended. “In the presence of severe renal failure...we suggest using unfractionated heparin followed by early vitamin K antagonists (possibly from day 1) or LMWH adjusted to anti-Xa concentration of the treatment of established VTE,” Dr. Farge and colleagues wrote.
Anticoagulant treatment is also recommended for a minimum of 3 months for children with symptomatic catheter-related thrombosis and as long as the central venous catheter is in place. For children with acute lymphoblastic leukemia who are undergoing induction chemotherapy, LMWH is also recommended as thromboprophylaxis.
For children who require a central venous catheter, the authors suggested that physicians use implanted ports over peripherally inserted central lines.
A version of this article first appeared on Medscape.com.
FROM THE LANCET ONCOLOGY
Registry launched to reach more patients with PF
The Pulmonary Fibrosis Foundation (PFF) has launched a new initiative in which they hope to capture a far more diverse representation of patients with pulmonary fibrosis (PF) than the current registry allows them to do, a press release from the PFF indicated.
“The existing registry we have – the PFF Patient Registry – is limited to our care centers, which are primarily academic clinical institutions and we have only a few thousand patients within that registry,” Junelle Speller, MBA, vice president of the PFF Registry, told this news organization.
“We wanted to go beyond these care centers and capture patients in community centers, and in rural settings to provide a more complete understanding of patients with this disease and, of course, have a larger sample size,” she added.
“ across all parts of the United States and, most importantly, accelerate the research on PF toward improving earlier diagnosis, treatment, and outcomes for these patients,” Ms. Speller said.
Passive versus active
The PFF Community Registry differs in its structure and purpose from the PFF Patient Registry, as Ms. Speller explained. First, the PFF Patient Registry, established in 2016, is “passive” in its nature in that whatever information is entered into a patient’s electronic medical record or clinical chart on a routine office visit is abstracted and captured in the registry. By contrast, the PFF Community Registry is asking for self-reported data from patients, “so it’s more of an ‘active’ registry and will give us a chance to have a bidirectional connection with participants, provide email updates and newsletters, and give patients an opportunity to participate in future studies within the registry as well as in clinical trials,” she noted.
The two registries still overlap in that both capture demographic data on patients’ medical and family histories as well as any medications patients may be taking, but the Community Registry will also capture information with respect to education, employment, patient-reported outcomes, and quality of life metrics. “It will also let us know how patients feel about continued education on the disease itself and patient participation in support groups,” Ms. Speller observed.
The Community Registry will also collect information from lung transplant recipients who have had PF or any other form of interstitial lung disease (ILD) as well as information from caregivers and family members affected by the patient’s disease. As Ms. Speller noted, both PF and other forms of ILD (of which there are more than 200 types) are all characterized by inflammation or scarring in the lung. “Patients are often misdiagnosed, and it can take months, even years, to identify the disease,” Ms. Speller said.
From there, it can be a very long and difficult road ahead, with no cure in sight, although several antifibrotic drugs do help slow disease progression. Typically, onset is around the age of 60 and symptoms include chronic dry cough, fatigue, shortness of breath, weakness, discomfort in the chest, and sometimes unexplained weight loss. Some patients do have a history of smoking, but not all, Ms. Speller noted. So far, registry data suggest PF largely occurs in White patients.
“We’re very excited about the Community Registry, particularly about reaching into communities that we haven’t been able to reach with our existing registry,” Ms. Speller noted. “The rural population in particular is often underserved, so we are really looking forward to capturing data from these patients as well as those from community centers within smaller and larger cities,” she observed.
“A powerful aspect of the Community Registry is that we can use the information gained from it to understand the experience of individuals living with PF, and how it impacts their lives and those of their families and caregivers,” Kevin Flaherty, MD, steering committee chair, PFF Registry, said in a statement.
“Researchers can also look at the data to better understand fibrotic lung diseases and learn about effective approaches to improve patient care,” he added.
Patients who wish to join the PFF Community Registry can sign up at pffregistry.org. To learn more about PF and its risk factors, readers are invited to visit www.AboutPF.org. More than 250,000 patients in the United States are living with either PF or other types of ILD.
Ms. Speller and Dr. Flaherty disclosed no financial conflicts of interest. The PFF Registry is supported by its founding partner, Genentech, Visionary Partner, United Therapeutics, and its sustaining partner, Boehringer Ingelheim, as well as many donors.
A version of this article first appeared on Medscape.com.
This article was updated 8/8/22.
The Pulmonary Fibrosis Foundation (PFF) has launched a new initiative in which they hope to capture a far more diverse representation of patients with pulmonary fibrosis (PF) than the current registry allows them to do, a press release from the PFF indicated.
“The existing registry we have – the PFF Patient Registry – is limited to our care centers, which are primarily academic clinical institutions and we have only a few thousand patients within that registry,” Junelle Speller, MBA, vice president of the PFF Registry, told this news organization.
“We wanted to go beyond these care centers and capture patients in community centers, and in rural settings to provide a more complete understanding of patients with this disease and, of course, have a larger sample size,” she added.
“ across all parts of the United States and, most importantly, accelerate the research on PF toward improving earlier diagnosis, treatment, and outcomes for these patients,” Ms. Speller said.
Passive versus active
The PFF Community Registry differs in its structure and purpose from the PFF Patient Registry, as Ms. Speller explained. First, the PFF Patient Registry, established in 2016, is “passive” in its nature in that whatever information is entered into a patient’s electronic medical record or clinical chart on a routine office visit is abstracted and captured in the registry. By contrast, the PFF Community Registry is asking for self-reported data from patients, “so it’s more of an ‘active’ registry and will give us a chance to have a bidirectional connection with participants, provide email updates and newsletters, and give patients an opportunity to participate in future studies within the registry as well as in clinical trials,” she noted.
The two registries still overlap in that both capture demographic data on patients’ medical and family histories as well as any medications patients may be taking, but the Community Registry will also capture information with respect to education, employment, patient-reported outcomes, and quality of life metrics. “It will also let us know how patients feel about continued education on the disease itself and patient participation in support groups,” Ms. Speller observed.
The Community Registry will also collect information from lung transplant recipients who have had PF or any other form of interstitial lung disease (ILD) as well as information from caregivers and family members affected by the patient’s disease. As Ms. Speller noted, both PF and other forms of ILD (of which there are more than 200 types) are all characterized by inflammation or scarring in the lung. “Patients are often misdiagnosed, and it can take months, even years, to identify the disease,” Ms. Speller said.
From there, it can be a very long and difficult road ahead, with no cure in sight, although several antifibrotic drugs do help slow disease progression. Typically, onset is around the age of 60 and symptoms include chronic dry cough, fatigue, shortness of breath, weakness, discomfort in the chest, and sometimes unexplained weight loss. Some patients do have a history of smoking, but not all, Ms. Speller noted. So far, registry data suggest PF largely occurs in White patients.
“We’re very excited about the Community Registry, particularly about reaching into communities that we haven’t been able to reach with our existing registry,” Ms. Speller noted. “The rural population in particular is often underserved, so we are really looking forward to capturing data from these patients as well as those from community centers within smaller and larger cities,” she observed.
“A powerful aspect of the Community Registry is that we can use the information gained from it to understand the experience of individuals living with PF, and how it impacts their lives and those of their families and caregivers,” Kevin Flaherty, MD, steering committee chair, PFF Registry, said in a statement.
“Researchers can also look at the data to better understand fibrotic lung diseases and learn about effective approaches to improve patient care,” he added.
Patients who wish to join the PFF Community Registry can sign up at pffregistry.org. To learn more about PF and its risk factors, readers are invited to visit www.AboutPF.org. More than 250,000 patients in the United States are living with either PF or other types of ILD.
Ms. Speller and Dr. Flaherty disclosed no financial conflicts of interest. The PFF Registry is supported by its founding partner, Genentech, Visionary Partner, United Therapeutics, and its sustaining partner, Boehringer Ingelheim, as well as many donors.
A version of this article first appeared on Medscape.com.
This article was updated 8/8/22.
The Pulmonary Fibrosis Foundation (PFF) has launched a new initiative in which they hope to capture a far more diverse representation of patients with pulmonary fibrosis (PF) than the current registry allows them to do, a press release from the PFF indicated.
“The existing registry we have – the PFF Patient Registry – is limited to our care centers, which are primarily academic clinical institutions and we have only a few thousand patients within that registry,” Junelle Speller, MBA, vice president of the PFF Registry, told this news organization.
“We wanted to go beyond these care centers and capture patients in community centers, and in rural settings to provide a more complete understanding of patients with this disease and, of course, have a larger sample size,” she added.
“ across all parts of the United States and, most importantly, accelerate the research on PF toward improving earlier diagnosis, treatment, and outcomes for these patients,” Ms. Speller said.
Passive versus active
The PFF Community Registry differs in its structure and purpose from the PFF Patient Registry, as Ms. Speller explained. First, the PFF Patient Registry, established in 2016, is “passive” in its nature in that whatever information is entered into a patient’s electronic medical record or clinical chart on a routine office visit is abstracted and captured in the registry. By contrast, the PFF Community Registry is asking for self-reported data from patients, “so it’s more of an ‘active’ registry and will give us a chance to have a bidirectional connection with participants, provide email updates and newsletters, and give patients an opportunity to participate in future studies within the registry as well as in clinical trials,” she noted.
The two registries still overlap in that both capture demographic data on patients’ medical and family histories as well as any medications patients may be taking, but the Community Registry will also capture information with respect to education, employment, patient-reported outcomes, and quality of life metrics. “It will also let us know how patients feel about continued education on the disease itself and patient participation in support groups,” Ms. Speller observed.
The Community Registry will also collect information from lung transplant recipients who have had PF or any other form of interstitial lung disease (ILD) as well as information from caregivers and family members affected by the patient’s disease. As Ms. Speller noted, both PF and other forms of ILD (of which there are more than 200 types) are all characterized by inflammation or scarring in the lung. “Patients are often misdiagnosed, and it can take months, even years, to identify the disease,” Ms. Speller said.
From there, it can be a very long and difficult road ahead, with no cure in sight, although several antifibrotic drugs do help slow disease progression. Typically, onset is around the age of 60 and symptoms include chronic dry cough, fatigue, shortness of breath, weakness, discomfort in the chest, and sometimes unexplained weight loss. Some patients do have a history of smoking, but not all, Ms. Speller noted. So far, registry data suggest PF largely occurs in White patients.
“We’re very excited about the Community Registry, particularly about reaching into communities that we haven’t been able to reach with our existing registry,” Ms. Speller noted. “The rural population in particular is often underserved, so we are really looking forward to capturing data from these patients as well as those from community centers within smaller and larger cities,” she observed.
“A powerful aspect of the Community Registry is that we can use the information gained from it to understand the experience of individuals living with PF, and how it impacts their lives and those of their families and caregivers,” Kevin Flaherty, MD, steering committee chair, PFF Registry, said in a statement.
“Researchers can also look at the data to better understand fibrotic lung diseases and learn about effective approaches to improve patient care,” he added.
Patients who wish to join the PFF Community Registry can sign up at pffregistry.org. To learn more about PF and its risk factors, readers are invited to visit www.AboutPF.org. More than 250,000 patients in the United States are living with either PF or other types of ILD.
Ms. Speller and Dr. Flaherty disclosed no financial conflicts of interest. The PFF Registry is supported by its founding partner, Genentech, Visionary Partner, United Therapeutics, and its sustaining partner, Boehringer Ingelheim, as well as many donors.
A version of this article first appeared on Medscape.com.
This article was updated 8/8/22.
Low calcium, potassium key risk factors for kidney stones
as well as their symptomatic recurrence, a population-based study of dietary factors shows.
“Our research is of particular importance as recommendations for preventing symptomatic recurrence of kidney stones has largely been based on dietary factors associated with the incidence rather than the recurrence of stone formation,” Api Chewcharat, MD, Mayo Clinic, Rochester, Minn., said in a video discussing the study.
“We recommend a daily intake of calcium of approximately 1,200 mg and a diet that is high in potassium, especially high in fruits and vegetables, in order to prevent both incident and recurrent symptomatic kidney stone formation,” he stressed.
The study was published online in Mayo Clinic Proceedings.
Lower dietary calcium, potassium, and fluid associated with increased incidence
Some 411 patients with incident symptomatic kidney stone formation were recruited. Diets were compared between them and 384 controls. Patients were seen at the Mayo Clinic in either Minnesota or Florida between Jan. 1, 2009, and Aug. 31, 2018. “Dietary factors were based on a Viocare food frequency questionnaire administered during a baseline in-person study visit,” Dr. Chewcharat and colleagues observed.
During a median follow-up of 4.1 years, 73 patients experienced a symptomatic recurrence. In a fully adjusted analysis, a dietary calcium intake less than 1,200 mg/d was associated with incident stone formation. Similarly, among participants with a fluid intake less than 3,400 mL/d – about nine 12-oz glasses of fluid – was also associated with incident stone formation, as was a lower intake of dietary potassium, caffeine, and phytate. Phytate is an antioxidant found in whole grains, nuts, and other foods that can increase calcium absorption and urinary calcium excretion.
After excluding patients who were taking either a thiazide diuretic or a calcium supplement, lower dietary calcium and potassium, fluid, and phytate intake remained significantly associated with incident stone formation.
However, only lower dietary calcium intake was associated with a higher risk for symptomatic recurrence, although a lower dietary potassium intake was also associated with a higher risk for symptomatic recurrence in an analysis that adjusted for body mass index, fluid, and energy intake.
As the authors suggested, patients may be less keen to adjust their diet to prevent the development of incident kidney stones. On the other hand, they may be much more willing to adjust their diet to prevent their symptomatic recurrence. The Department of Agriculture currently recommends that individuals get approximately 1,200 mg/d of dietary calcium which, given the study results, appears to be justified for the prevention of symptomatic stone recurrence.
A higher-calcium diet is associated with a higher urinary pH, and citrate confers an alkali load which helps protect against the formation of calcium oxalate stones. Foods that are high in potassium also contain more fluid, citrate, and phytate, which, again, have been reported to be protective against kidney stones. “Changing your diet to prevent kidney stones can be very difficult,” Andrew Rule, MD, a nephrologist at the Mayo Clinic said in a statement.
“Thus, knowing the dietary factors that are most important for preventing kidney stone recurrence can help patients and providers know what to prioritize,” he added.
The authors have no conflicts of interest to declare.
A version of this article first appeared on Medscape.com.
as well as their symptomatic recurrence, a population-based study of dietary factors shows.
“Our research is of particular importance as recommendations for preventing symptomatic recurrence of kidney stones has largely been based on dietary factors associated with the incidence rather than the recurrence of stone formation,” Api Chewcharat, MD, Mayo Clinic, Rochester, Minn., said in a video discussing the study.
“We recommend a daily intake of calcium of approximately 1,200 mg and a diet that is high in potassium, especially high in fruits and vegetables, in order to prevent both incident and recurrent symptomatic kidney stone formation,” he stressed.
The study was published online in Mayo Clinic Proceedings.
Lower dietary calcium, potassium, and fluid associated with increased incidence
Some 411 patients with incident symptomatic kidney stone formation were recruited. Diets were compared between them and 384 controls. Patients were seen at the Mayo Clinic in either Minnesota or Florida between Jan. 1, 2009, and Aug. 31, 2018. “Dietary factors were based on a Viocare food frequency questionnaire administered during a baseline in-person study visit,” Dr. Chewcharat and colleagues observed.
During a median follow-up of 4.1 years, 73 patients experienced a symptomatic recurrence. In a fully adjusted analysis, a dietary calcium intake less than 1,200 mg/d was associated with incident stone formation. Similarly, among participants with a fluid intake less than 3,400 mL/d – about nine 12-oz glasses of fluid – was also associated with incident stone formation, as was a lower intake of dietary potassium, caffeine, and phytate. Phytate is an antioxidant found in whole grains, nuts, and other foods that can increase calcium absorption and urinary calcium excretion.
After excluding patients who were taking either a thiazide diuretic or a calcium supplement, lower dietary calcium and potassium, fluid, and phytate intake remained significantly associated with incident stone formation.
However, only lower dietary calcium intake was associated with a higher risk for symptomatic recurrence, although a lower dietary potassium intake was also associated with a higher risk for symptomatic recurrence in an analysis that adjusted for body mass index, fluid, and energy intake.
As the authors suggested, patients may be less keen to adjust their diet to prevent the development of incident kidney stones. On the other hand, they may be much more willing to adjust their diet to prevent their symptomatic recurrence. The Department of Agriculture currently recommends that individuals get approximately 1,200 mg/d of dietary calcium which, given the study results, appears to be justified for the prevention of symptomatic stone recurrence.
A higher-calcium diet is associated with a higher urinary pH, and citrate confers an alkali load which helps protect against the formation of calcium oxalate stones. Foods that are high in potassium also contain more fluid, citrate, and phytate, which, again, have been reported to be protective against kidney stones. “Changing your diet to prevent kidney stones can be very difficult,” Andrew Rule, MD, a nephrologist at the Mayo Clinic said in a statement.
“Thus, knowing the dietary factors that are most important for preventing kidney stone recurrence can help patients and providers know what to prioritize,” he added.
The authors have no conflicts of interest to declare.
A version of this article first appeared on Medscape.com.
as well as their symptomatic recurrence, a population-based study of dietary factors shows.
“Our research is of particular importance as recommendations for preventing symptomatic recurrence of kidney stones has largely been based on dietary factors associated with the incidence rather than the recurrence of stone formation,” Api Chewcharat, MD, Mayo Clinic, Rochester, Minn., said in a video discussing the study.
“We recommend a daily intake of calcium of approximately 1,200 mg and a diet that is high in potassium, especially high in fruits and vegetables, in order to prevent both incident and recurrent symptomatic kidney stone formation,” he stressed.
The study was published online in Mayo Clinic Proceedings.
Lower dietary calcium, potassium, and fluid associated with increased incidence
Some 411 patients with incident symptomatic kidney stone formation were recruited. Diets were compared between them and 384 controls. Patients were seen at the Mayo Clinic in either Minnesota or Florida between Jan. 1, 2009, and Aug. 31, 2018. “Dietary factors were based on a Viocare food frequency questionnaire administered during a baseline in-person study visit,” Dr. Chewcharat and colleagues observed.
During a median follow-up of 4.1 years, 73 patients experienced a symptomatic recurrence. In a fully adjusted analysis, a dietary calcium intake less than 1,200 mg/d was associated with incident stone formation. Similarly, among participants with a fluid intake less than 3,400 mL/d – about nine 12-oz glasses of fluid – was also associated with incident stone formation, as was a lower intake of dietary potassium, caffeine, and phytate. Phytate is an antioxidant found in whole grains, nuts, and other foods that can increase calcium absorption and urinary calcium excretion.
After excluding patients who were taking either a thiazide diuretic or a calcium supplement, lower dietary calcium and potassium, fluid, and phytate intake remained significantly associated with incident stone formation.
However, only lower dietary calcium intake was associated with a higher risk for symptomatic recurrence, although a lower dietary potassium intake was also associated with a higher risk for symptomatic recurrence in an analysis that adjusted for body mass index, fluid, and energy intake.
As the authors suggested, patients may be less keen to adjust their diet to prevent the development of incident kidney stones. On the other hand, they may be much more willing to adjust their diet to prevent their symptomatic recurrence. The Department of Agriculture currently recommends that individuals get approximately 1,200 mg/d of dietary calcium which, given the study results, appears to be justified for the prevention of symptomatic stone recurrence.
A higher-calcium diet is associated with a higher urinary pH, and citrate confers an alkali load which helps protect against the formation of calcium oxalate stones. Foods that are high in potassium also contain more fluid, citrate, and phytate, which, again, have been reported to be protective against kidney stones. “Changing your diet to prevent kidney stones can be very difficult,” Andrew Rule, MD, a nephrologist at the Mayo Clinic said in a statement.
“Thus, knowing the dietary factors that are most important for preventing kidney stone recurrence can help patients and providers know what to prioritize,” he added.
The authors have no conflicts of interest to declare.
A version of this article first appeared on Medscape.com.
FROM MAYO CLINIC PRECEEDINGS
Steroids no cure for obstructive sleep apnea in children
Children with obstructive sleep apnea syndrome (OSAS) who undergo treatment with intranasal corticosteroids (INCS) did not experience significant improvement in polysomnographic, neurobehavioral, and other symptoms at 3 and 12 months of treatment. At 12 months of INCS treatment, there was a statistically significant but not clinically relevant reduction in the obstructive apnea hypopnea index (OHAI).
“Previous studies were done in children with OSA with an obstructive apnea hypopnea index of less than 5, so they had very mild OSA,” Ignacio Tapia, MD, associate professor of pediatrics, University of Pennsylvania, Philadelphia, said in an interview.
“But then people started using the INCS for a whole range of OSA, so this is why we wanted to do the trial, to make sure that these drugs were being used correctly,” he added.
“The main message from this paper is that I think INCS may still have a role to play in OSA to treat some of the symptoms, like snoring, and in our study, quality of life indices also improved, ,” Dr. Tapia emphasized.
The study was published online in the journal Chest.
3 months of INCS
A total of 134 children between 5 and 12 years of age were randomly assigned to receive INCS for 3 months or placebo. Children in the original INCS arm were then reassigned to receive 9 more months of the same treatment or placebo. Symptoms as well as polysomnographic and neurobehavioral findings were measured at baseline, at 3 months, and again at 12 months.
“The primary outcome was OAHI change at 3 months, available for 122 children,” the authors explained. The OSAS was defined as an OAHI of between two and three events per hour. The median age of the children at baseline was 7.9 years, and the median OAHI baseline score was 5.8/hr (95% confidence interval, 3.6-9.7/hr). The total daily dose of the INCS used was 110 mcg.
At 3 months, the mean change in the OAHI from baseline was –1.73/hr (95% CI, –3.91 to 1.92/hr), while at 12 months, the mean change in the same index was –1.21 (–4.22 to 1.71/hr). These changes were not significantly different from OAHI changes observed among control participants. “OSAS symptoms and neurobehavioral results were not different [either] between the INCS and placebo groups at 3 and12 months,” the authors added.
However, among those children who received INCS treatment for the entire 12 months, the OAHI decreased significantly from 7.2/hr (95% CI, 3.62-9.88/hr) at baseline to 3.71/hr (95% CI, 1.56-6.4/hr; P = .039), although the OAHI did not normalize, the authors noted. Asked to clarify whether this change was not significant, Dr. Tapia said that it did meet statistical significance, but clinically, it meant that the children still needed some form of treatment, because they still had OSA in the range needing treatment.
The placebo group had more asthma exacerbations, upper respiratory tract infections, and exacerbations of OSAS symptoms, compared with children in the INCS group. It is possible that INCS provided a certain degree of protection from asthma exacerbation, the authors suggested.
However, recent guidelines from the American Academy of Pediatrics suggest that clinicians may prescribe these agents for children with mild OSAS in whom adenotonsillectomy is contraindicated; for those with mild postoperative OSAS, adenotonsillectomy remains the treatment of choice for childhood OSA. “The low level of enthusiasm for INCS in these guidelines is based on results from studies of INCS treatment of OSAS that had been limited by small sample size, lack of placebo control, limited duration, and variability in baseline data,” the authors wrote.
“The results of the current larger and more rigorous study of children with a wider range of OSAS also do not support the currently liberal use of INCS for the treatment of OSAS,” they wrote.
Complex issue
In a comment, Rakesh Bhattacharjee, MD, associate professor of pediatrics, University of California, San Diego, noted that he does prescribe INCS for children with mild OSA but not for all children. “We based our decisions on polysomnography, which we use to categorize OSA as mild, moderate, or severe.
“But we certainly do offer this treatment for children with mild sleep apnea as a way to avoid surgical treatment,” Dr. Bhattacharjee added. He also uses INCS for residual sleep apnea that some children experience following adenotonsillectomy. As the current study suggests, many people are treating sleep apnea empirically without confirming the severity of the disorder by a sleep study.
“If a sleep study is not done, we don’t know how severe it is, so this would advocate for the utility of a sleep study so that you can quantify the severity of symptoms and target your therapy to children who might be appropriate for INCS therapy,” Dr. Bhattacharjee said.
On the other hand, surgery is not always relevant even if a child has enlarged adenoids and tonsils, as, for example, a child with obesity. In these children, physicians need to think about other treatments, such as continuous positive airways pressure. “CPAP is not perfect,” Dr. Bhattacharjee observed. “And as pediatricians, we need to do a lot of work to improve the use of CPAP, but, that said, there are children for whom INCS and surgery might be a waste of time, and this is where CPAP might be an alternative.”
Dr. Bhattacharjee previously was the lead author of a large study of children who underwent treatment with CPAP. While findings suggested that adherence to treatment is lower in children than it is for adults, the authors also showed that numerous actionable factors could used to improve adherence to CPAP among children who might otherwise benefit from it.
The authors disclosed no relevant financial relationships. Dr. Bhattacharjee has served as a scientific adviser for Jazz Pharmaceuticals.
A version of this article first appeared on Medscape.com.
Children with obstructive sleep apnea syndrome (OSAS) who undergo treatment with intranasal corticosteroids (INCS) did not experience significant improvement in polysomnographic, neurobehavioral, and other symptoms at 3 and 12 months of treatment. At 12 months of INCS treatment, there was a statistically significant but not clinically relevant reduction in the obstructive apnea hypopnea index (OHAI).
“Previous studies were done in children with OSA with an obstructive apnea hypopnea index of less than 5, so they had very mild OSA,” Ignacio Tapia, MD, associate professor of pediatrics, University of Pennsylvania, Philadelphia, said in an interview.
“But then people started using the INCS for a whole range of OSA, so this is why we wanted to do the trial, to make sure that these drugs were being used correctly,” he added.
“The main message from this paper is that I think INCS may still have a role to play in OSA to treat some of the symptoms, like snoring, and in our study, quality of life indices also improved, ,” Dr. Tapia emphasized.
The study was published online in the journal Chest.
3 months of INCS
A total of 134 children between 5 and 12 years of age were randomly assigned to receive INCS for 3 months or placebo. Children in the original INCS arm were then reassigned to receive 9 more months of the same treatment or placebo. Symptoms as well as polysomnographic and neurobehavioral findings were measured at baseline, at 3 months, and again at 12 months.
“The primary outcome was OAHI change at 3 months, available for 122 children,” the authors explained. The OSAS was defined as an OAHI of between two and three events per hour. The median age of the children at baseline was 7.9 years, and the median OAHI baseline score was 5.8/hr (95% confidence interval, 3.6-9.7/hr). The total daily dose of the INCS used was 110 mcg.
At 3 months, the mean change in the OAHI from baseline was –1.73/hr (95% CI, –3.91 to 1.92/hr), while at 12 months, the mean change in the same index was –1.21 (–4.22 to 1.71/hr). These changes were not significantly different from OAHI changes observed among control participants. “OSAS symptoms and neurobehavioral results were not different [either] between the INCS and placebo groups at 3 and12 months,” the authors added.
However, among those children who received INCS treatment for the entire 12 months, the OAHI decreased significantly from 7.2/hr (95% CI, 3.62-9.88/hr) at baseline to 3.71/hr (95% CI, 1.56-6.4/hr; P = .039), although the OAHI did not normalize, the authors noted. Asked to clarify whether this change was not significant, Dr. Tapia said that it did meet statistical significance, but clinically, it meant that the children still needed some form of treatment, because they still had OSA in the range needing treatment.
The placebo group had more asthma exacerbations, upper respiratory tract infections, and exacerbations of OSAS symptoms, compared with children in the INCS group. It is possible that INCS provided a certain degree of protection from asthma exacerbation, the authors suggested.
However, recent guidelines from the American Academy of Pediatrics suggest that clinicians may prescribe these agents for children with mild OSAS in whom adenotonsillectomy is contraindicated; for those with mild postoperative OSAS, adenotonsillectomy remains the treatment of choice for childhood OSA. “The low level of enthusiasm for INCS in these guidelines is based on results from studies of INCS treatment of OSAS that had been limited by small sample size, lack of placebo control, limited duration, and variability in baseline data,” the authors wrote.
“The results of the current larger and more rigorous study of children with a wider range of OSAS also do not support the currently liberal use of INCS for the treatment of OSAS,” they wrote.
Complex issue
In a comment, Rakesh Bhattacharjee, MD, associate professor of pediatrics, University of California, San Diego, noted that he does prescribe INCS for children with mild OSA but not for all children. “We based our decisions on polysomnography, which we use to categorize OSA as mild, moderate, or severe.
“But we certainly do offer this treatment for children with mild sleep apnea as a way to avoid surgical treatment,” Dr. Bhattacharjee added. He also uses INCS for residual sleep apnea that some children experience following adenotonsillectomy. As the current study suggests, many people are treating sleep apnea empirically without confirming the severity of the disorder by a sleep study.
“If a sleep study is not done, we don’t know how severe it is, so this would advocate for the utility of a sleep study so that you can quantify the severity of symptoms and target your therapy to children who might be appropriate for INCS therapy,” Dr. Bhattacharjee said.
On the other hand, surgery is not always relevant even if a child has enlarged adenoids and tonsils, as, for example, a child with obesity. In these children, physicians need to think about other treatments, such as continuous positive airways pressure. “CPAP is not perfect,” Dr. Bhattacharjee observed. “And as pediatricians, we need to do a lot of work to improve the use of CPAP, but, that said, there are children for whom INCS and surgery might be a waste of time, and this is where CPAP might be an alternative.”
Dr. Bhattacharjee previously was the lead author of a large study of children who underwent treatment with CPAP. While findings suggested that adherence to treatment is lower in children than it is for adults, the authors also showed that numerous actionable factors could used to improve adherence to CPAP among children who might otherwise benefit from it.
The authors disclosed no relevant financial relationships. Dr. Bhattacharjee has served as a scientific adviser for Jazz Pharmaceuticals.
A version of this article first appeared on Medscape.com.
Children with obstructive sleep apnea syndrome (OSAS) who undergo treatment with intranasal corticosteroids (INCS) did not experience significant improvement in polysomnographic, neurobehavioral, and other symptoms at 3 and 12 months of treatment. At 12 months of INCS treatment, there was a statistically significant but not clinically relevant reduction in the obstructive apnea hypopnea index (OHAI).
“Previous studies were done in children with OSA with an obstructive apnea hypopnea index of less than 5, so they had very mild OSA,” Ignacio Tapia, MD, associate professor of pediatrics, University of Pennsylvania, Philadelphia, said in an interview.
“But then people started using the INCS for a whole range of OSA, so this is why we wanted to do the trial, to make sure that these drugs were being used correctly,” he added.
“The main message from this paper is that I think INCS may still have a role to play in OSA to treat some of the symptoms, like snoring, and in our study, quality of life indices also improved, ,” Dr. Tapia emphasized.
The study was published online in the journal Chest.
3 months of INCS
A total of 134 children between 5 and 12 years of age were randomly assigned to receive INCS for 3 months or placebo. Children in the original INCS arm were then reassigned to receive 9 more months of the same treatment or placebo. Symptoms as well as polysomnographic and neurobehavioral findings were measured at baseline, at 3 months, and again at 12 months.
“The primary outcome was OAHI change at 3 months, available for 122 children,” the authors explained. The OSAS was defined as an OAHI of between two and three events per hour. The median age of the children at baseline was 7.9 years, and the median OAHI baseline score was 5.8/hr (95% confidence interval, 3.6-9.7/hr). The total daily dose of the INCS used was 110 mcg.
At 3 months, the mean change in the OAHI from baseline was –1.73/hr (95% CI, –3.91 to 1.92/hr), while at 12 months, the mean change in the same index was –1.21 (–4.22 to 1.71/hr). These changes were not significantly different from OAHI changes observed among control participants. “OSAS symptoms and neurobehavioral results were not different [either] between the INCS and placebo groups at 3 and12 months,” the authors added.
However, among those children who received INCS treatment for the entire 12 months, the OAHI decreased significantly from 7.2/hr (95% CI, 3.62-9.88/hr) at baseline to 3.71/hr (95% CI, 1.56-6.4/hr; P = .039), although the OAHI did not normalize, the authors noted. Asked to clarify whether this change was not significant, Dr. Tapia said that it did meet statistical significance, but clinically, it meant that the children still needed some form of treatment, because they still had OSA in the range needing treatment.
The placebo group had more asthma exacerbations, upper respiratory tract infections, and exacerbations of OSAS symptoms, compared with children in the INCS group. It is possible that INCS provided a certain degree of protection from asthma exacerbation, the authors suggested.
However, recent guidelines from the American Academy of Pediatrics suggest that clinicians may prescribe these agents for children with mild OSAS in whom adenotonsillectomy is contraindicated; for those with mild postoperative OSAS, adenotonsillectomy remains the treatment of choice for childhood OSA. “The low level of enthusiasm for INCS in these guidelines is based on results from studies of INCS treatment of OSAS that had been limited by small sample size, lack of placebo control, limited duration, and variability in baseline data,” the authors wrote.
“The results of the current larger and more rigorous study of children with a wider range of OSAS also do not support the currently liberal use of INCS for the treatment of OSAS,” they wrote.
Complex issue
In a comment, Rakesh Bhattacharjee, MD, associate professor of pediatrics, University of California, San Diego, noted that he does prescribe INCS for children with mild OSA but not for all children. “We based our decisions on polysomnography, which we use to categorize OSA as mild, moderate, or severe.
“But we certainly do offer this treatment for children with mild sleep apnea as a way to avoid surgical treatment,” Dr. Bhattacharjee added. He also uses INCS for residual sleep apnea that some children experience following adenotonsillectomy. As the current study suggests, many people are treating sleep apnea empirically without confirming the severity of the disorder by a sleep study.
“If a sleep study is not done, we don’t know how severe it is, so this would advocate for the utility of a sleep study so that you can quantify the severity of symptoms and target your therapy to children who might be appropriate for INCS therapy,” Dr. Bhattacharjee said.
On the other hand, surgery is not always relevant even if a child has enlarged adenoids and tonsils, as, for example, a child with obesity. In these children, physicians need to think about other treatments, such as continuous positive airways pressure. “CPAP is not perfect,” Dr. Bhattacharjee observed. “And as pediatricians, we need to do a lot of work to improve the use of CPAP, but, that said, there are children for whom INCS and surgery might be a waste of time, and this is where CPAP might be an alternative.”
Dr. Bhattacharjee previously was the lead author of a large study of children who underwent treatment with CPAP. While findings suggested that adherence to treatment is lower in children than it is for adults, the authors also showed that numerous actionable factors could used to improve adherence to CPAP among children who might otherwise benefit from it.
The authors disclosed no relevant financial relationships. Dr. Bhattacharjee has served as a scientific adviser for Jazz Pharmaceuticals.
A version of this article first appeared on Medscape.com.
FROM CHEST
Minimal differences between biologics approved for severe asthma
Differences in the safety and efficacy between the biologics approved for the treatment of severe eosinophilic asthma are so minimal as to not meet clinically important thresholds, a network meta-analysis shows.
“We know relatively little of the comparative effectiveness or safety of biologics approved for the treatment of asthma [but since] Ayobami Akenroye, MD, MPH, of Brigham and Women’s Hospital and Harvard Medical School, both in Boston, said in an interview.
“But the decision to use one biologic or not is complex and goes beyond comparative effectiveness, and factors such as insurance coverage, convenience of self-administration, and comorbidities all play a role in the choice of biologics,” she said, adding that all the outcomes assessed in the study contribute to or reflect a patient’s underlying asthma control.
The study was published online in the Journal of Allergy and Clinical Immunology.
Interleukin pathways
Drugs that target various interleukin signaling pathways involved in the pathogenesis of asthma include mepolizumab (Nucala), benralizumab (Fasenra), and dupilumab (Dupixent), all of which have been shown to decrease exacerbation rates, improve lung function, and enhance quality of life for patients with severe eosinophilic asthma. In a Bayesian network meta-analysis that allows for simultaneous comparisons of these three treatments, investigators analyzed eight randomized, placebo-controlled trials that compared each of the drugs with placebo. In total, the trials involved 6,461 patients; the duration of follow-up was between 24 and 56 weeks.
“In the subgroup of patients with eosinophil counts of ≥ 300 cells/mcL, all three biologics were significantly better than placebo in reducing exacerbations,” Dr. Akenroye and colleagues reported. For example, dupilumab reduced the exacerbation risk by 68% at a risk ratio of 0.32 (95% confidence interval, 0.23-0.45), while mepolizumab reduced it by almost as much at 63% (RR, 0.37; 95% CI, 0.30-0.45).
Benralizumab was slightly less effective than the other two biologics, reducing exacerbation risk by 51% (RR, 0.49; 95% CI, 0.43-0.55). “In patients with eosinophil counts of ≥ 300 cells/mcL, all three biologics had a probability of 1 in improving the exacerbation rate by 20% or more ... in comparison to placebo,” the authors wrote.
Regarding each drug’s effect in improving forced expiratory volume in 1 second (FEV1), the mean difference in milliliters with dupilumab before and after treatment was 230 (95% CI, 160-300), while for benralizumab, the MD was 150 (95% CI, 100-220) before and after treatment. With mepolizumab, the MD in FEV1 before and after treatment was also 150. In the same subgroup of patients with eosinophil counts of at least300 cells/mcL, all three biologics again had a probability of 1 in improving FEV1 by 50 mL or more above the placebo effect. A third endpoint that was analyzed was the potential reduction in asthma control questionnaire (ACQ) scores. With mepolizumab, the MD before and after treatment was –0.65 (95% CI, –0.81 to –0.45); with dupilumab, it was –0.48 (95% CI, –0.83 to –0.14); and with dupilumab, it was –0.32 (95% CI, –0.43 to –0.21).
“Dupilumab was significantly better than benralizumab in improving exacerbations,” the authors noted (RR, 0.66; 95% CI, 0.47-0.94), while mepolizumab was also better than benralizumab (RR, 0.75; 95% CI, 0.60-0.95). On the other hand, both dupilumab and benralizumab led to greater improvements in FEV1 than mepolizumab, although the effects of dupilumab and benralizumab on ACQ scores were not significantly different for patients whose lower eosinophil counts were between 150 and 299 cells/mcL.
As for safety outcomes, both mepolizumab and benralizumab were associated with a lower risk of serious adverse events, but dupilumab was not different from placebo in terms of overall safety, according to the authors. “The ultimate choice of biologic for each patient would ... depend on multiple factors including cost considerations and timing of administration.
“[However], these results may be helpful to clinicians as they optimize patient care,” they concluded. Limitations to the analysis include the fact that indirect comparisons cannot replace randomized trials that compare the three drugs directly.
It’s estimated that 5%-10% of the 26 million individuals with asthma in the United States have severe disease.
Dr. Akenroye disclosed no relevant financial relationships.
A version of this article first appeared on Medscape.com.
Differences in the safety and efficacy between the biologics approved for the treatment of severe eosinophilic asthma are so minimal as to not meet clinically important thresholds, a network meta-analysis shows.
“We know relatively little of the comparative effectiveness or safety of biologics approved for the treatment of asthma [but since] Ayobami Akenroye, MD, MPH, of Brigham and Women’s Hospital and Harvard Medical School, both in Boston, said in an interview.
“But the decision to use one biologic or not is complex and goes beyond comparative effectiveness, and factors such as insurance coverage, convenience of self-administration, and comorbidities all play a role in the choice of biologics,” she said, adding that all the outcomes assessed in the study contribute to or reflect a patient’s underlying asthma control.
The study was published online in the Journal of Allergy and Clinical Immunology.
Interleukin pathways
Drugs that target various interleukin signaling pathways involved in the pathogenesis of asthma include mepolizumab (Nucala), benralizumab (Fasenra), and dupilumab (Dupixent), all of which have been shown to decrease exacerbation rates, improve lung function, and enhance quality of life for patients with severe eosinophilic asthma. In a Bayesian network meta-analysis that allows for simultaneous comparisons of these three treatments, investigators analyzed eight randomized, placebo-controlled trials that compared each of the drugs with placebo. In total, the trials involved 6,461 patients; the duration of follow-up was between 24 and 56 weeks.
“In the subgroup of patients with eosinophil counts of ≥ 300 cells/mcL, all three biologics were significantly better than placebo in reducing exacerbations,” Dr. Akenroye and colleagues reported. For example, dupilumab reduced the exacerbation risk by 68% at a risk ratio of 0.32 (95% confidence interval, 0.23-0.45), while mepolizumab reduced it by almost as much at 63% (RR, 0.37; 95% CI, 0.30-0.45).
Benralizumab was slightly less effective than the other two biologics, reducing exacerbation risk by 51% (RR, 0.49; 95% CI, 0.43-0.55). “In patients with eosinophil counts of ≥ 300 cells/mcL, all three biologics had a probability of 1 in improving the exacerbation rate by 20% or more ... in comparison to placebo,” the authors wrote.
Regarding each drug’s effect in improving forced expiratory volume in 1 second (FEV1), the mean difference in milliliters with dupilumab before and after treatment was 230 (95% CI, 160-300), while for benralizumab, the MD was 150 (95% CI, 100-220) before and after treatment. With mepolizumab, the MD in FEV1 before and after treatment was also 150. In the same subgroup of patients with eosinophil counts of at least300 cells/mcL, all three biologics again had a probability of 1 in improving FEV1 by 50 mL or more above the placebo effect. A third endpoint that was analyzed was the potential reduction in asthma control questionnaire (ACQ) scores. With mepolizumab, the MD before and after treatment was –0.65 (95% CI, –0.81 to –0.45); with dupilumab, it was –0.48 (95% CI, –0.83 to –0.14); and with dupilumab, it was –0.32 (95% CI, –0.43 to –0.21).
“Dupilumab was significantly better than benralizumab in improving exacerbations,” the authors noted (RR, 0.66; 95% CI, 0.47-0.94), while mepolizumab was also better than benralizumab (RR, 0.75; 95% CI, 0.60-0.95). On the other hand, both dupilumab and benralizumab led to greater improvements in FEV1 than mepolizumab, although the effects of dupilumab and benralizumab on ACQ scores were not significantly different for patients whose lower eosinophil counts were between 150 and 299 cells/mcL.
As for safety outcomes, both mepolizumab and benralizumab were associated with a lower risk of serious adverse events, but dupilumab was not different from placebo in terms of overall safety, according to the authors. “The ultimate choice of biologic for each patient would ... depend on multiple factors including cost considerations and timing of administration.
“[However], these results may be helpful to clinicians as they optimize patient care,” they concluded. Limitations to the analysis include the fact that indirect comparisons cannot replace randomized trials that compare the three drugs directly.
It’s estimated that 5%-10% of the 26 million individuals with asthma in the United States have severe disease.
Dr. Akenroye disclosed no relevant financial relationships.
A version of this article first appeared on Medscape.com.
Differences in the safety and efficacy between the biologics approved for the treatment of severe eosinophilic asthma are so minimal as to not meet clinically important thresholds, a network meta-analysis shows.
“We know relatively little of the comparative effectiveness or safety of biologics approved for the treatment of asthma [but since] Ayobami Akenroye, MD, MPH, of Brigham and Women’s Hospital and Harvard Medical School, both in Boston, said in an interview.
“But the decision to use one biologic or not is complex and goes beyond comparative effectiveness, and factors such as insurance coverage, convenience of self-administration, and comorbidities all play a role in the choice of biologics,” she said, adding that all the outcomes assessed in the study contribute to or reflect a patient’s underlying asthma control.
The study was published online in the Journal of Allergy and Clinical Immunology.
Interleukin pathways
Drugs that target various interleukin signaling pathways involved in the pathogenesis of asthma include mepolizumab (Nucala), benralizumab (Fasenra), and dupilumab (Dupixent), all of which have been shown to decrease exacerbation rates, improve lung function, and enhance quality of life for patients with severe eosinophilic asthma. In a Bayesian network meta-analysis that allows for simultaneous comparisons of these three treatments, investigators analyzed eight randomized, placebo-controlled trials that compared each of the drugs with placebo. In total, the trials involved 6,461 patients; the duration of follow-up was between 24 and 56 weeks.
“In the subgroup of patients with eosinophil counts of ≥ 300 cells/mcL, all three biologics were significantly better than placebo in reducing exacerbations,” Dr. Akenroye and colleagues reported. For example, dupilumab reduced the exacerbation risk by 68% at a risk ratio of 0.32 (95% confidence interval, 0.23-0.45), while mepolizumab reduced it by almost as much at 63% (RR, 0.37; 95% CI, 0.30-0.45).
Benralizumab was slightly less effective than the other two biologics, reducing exacerbation risk by 51% (RR, 0.49; 95% CI, 0.43-0.55). “In patients with eosinophil counts of ≥ 300 cells/mcL, all three biologics had a probability of 1 in improving the exacerbation rate by 20% or more ... in comparison to placebo,” the authors wrote.
Regarding each drug’s effect in improving forced expiratory volume in 1 second (FEV1), the mean difference in milliliters with dupilumab before and after treatment was 230 (95% CI, 160-300), while for benralizumab, the MD was 150 (95% CI, 100-220) before and after treatment. With mepolizumab, the MD in FEV1 before and after treatment was also 150. In the same subgroup of patients with eosinophil counts of at least300 cells/mcL, all three biologics again had a probability of 1 in improving FEV1 by 50 mL or more above the placebo effect. A third endpoint that was analyzed was the potential reduction in asthma control questionnaire (ACQ) scores. With mepolizumab, the MD before and after treatment was –0.65 (95% CI, –0.81 to –0.45); with dupilumab, it was –0.48 (95% CI, –0.83 to –0.14); and with dupilumab, it was –0.32 (95% CI, –0.43 to –0.21).
“Dupilumab was significantly better than benralizumab in improving exacerbations,” the authors noted (RR, 0.66; 95% CI, 0.47-0.94), while mepolizumab was also better than benralizumab (RR, 0.75; 95% CI, 0.60-0.95). On the other hand, both dupilumab and benralizumab led to greater improvements in FEV1 than mepolizumab, although the effects of dupilumab and benralizumab on ACQ scores were not significantly different for patients whose lower eosinophil counts were between 150 and 299 cells/mcL.
As for safety outcomes, both mepolizumab and benralizumab were associated with a lower risk of serious adverse events, but dupilumab was not different from placebo in terms of overall safety, according to the authors. “The ultimate choice of biologic for each patient would ... depend on multiple factors including cost considerations and timing of administration.
“[However], these results may be helpful to clinicians as they optimize patient care,” they concluded. Limitations to the analysis include the fact that indirect comparisons cannot replace randomized trials that compare the three drugs directly.
It’s estimated that 5%-10% of the 26 million individuals with asthma in the United States have severe disease.
Dr. Akenroye disclosed no relevant financial relationships.
A version of this article first appeared on Medscape.com.
FROM THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY
Progression from nonradiographic to radiographic axial spondyloarthritis evaluated in multinational study
Four risk factors predicted the progression from nonradiographic to radiographic axial spondyloarthritis (axSpA) over a 5-year period in the PROOF study, a global, real-world, prospective, observational study carried out in 29 countries across six different geographic regions.
The predictors of progression within 5 years, based on the presence of radiographic sacroiliitis, included male gender, fulfillment of imaging criteria, HLA-B27 positivity, and a good response to NSAIDs, Denis Poddubnyy, MD, professor of rheumatology at Charité-Universitätsmedizin Berlin, said in his presentation of the study results at the annual European Congress of Rheumatology.
“In this study, 16% of nonradiographic axSpA patients progressed to radiographic axSpA within 5 years, with the mean time to disease progression of 2.4 years,” he said. PROOF (Patients with Axial Spondyloarthritis: Multicountry Registry of Clinical Characteristics) was originally designed to compare demographic and clinical characteristics of patients with axSpA across geographic regions.
In this particular analysis, Dr. Poddubnyy and colleagues aimed to track structural damage progression in the sacroiliac joint over time, as he explained in an interview. The study enrolled 2,633 adults with chronic back pain lasting for at least 3 months with onset before the age of 45 years. This analysis included patients diagnosed with axSpA who also fulfilled the Assessment of SpondyloArthritis international Society (ASAS) classification criteria for axSpA.
Both baseline and follow-up radiographs of sacroiliac joints were evaluated for those with an initial diagnosis of nonradiographic axSpA by two central readers; in cases when the readers disagreed on the classification – either nonradiographic or radiographic axSpA – images were adjudicated by a third reader. Radiographic progression from nonradiographic to radiographic axSpA was evaluated over the next 5 years.
Among all enrolled patients, 82% (n = 2,165) were diagnosed with axSpA and fulfilled the ASAS classification criteria. Of 1,612 who were classified by central reading, 65% had radiographic axSpA while the remaining 35% had nonradiographic axSpA. About 78% of those with nonradiographic axSpA fulfilled the ASAS classification criteria because of positive findings on imaging plus one or more features of spondyloarthritis. The other 22% were classified according to clinical criteria.
A total of 246 nonradiographic axSpA patients who had one or more follow-up radiographs of the sacroiliac joint were included in the current analysis. In this smaller group of patients, progression from the initial diagnosis of nonradiographic to radiographic axSpA at any one point over the 5-year follow-up occurred in 40 patients (16%) at a range of between 0.9 and 5.1 years.
“Females are more likely to stay in the nonradiographic stage than males,” Dr. Poddubnyy noted. Indeed, male gender conferred an over-threefold higher risk of radiographic progression, compared with females (hazard ratio, 3.16; 95% confidence interval, 1.22-8.17; P = .0174). Fulfillment of imaging criteria – in other words, the presence of inflammation on MRI – was also a strong predictor of progression, conferring an over-sixfold risk of radiographic progression (HR, 6.64; 95% CI, 1.37-32.25; P = .0188).
Interestingly, a good response to NSAIDs – the mainstay treatment for both nonradiographic and radiographic axSpA – was also significantly associated with radiographic progression, conferring an over-fourfold risk among those with an initial diagnosis of nonradiographic axSpA (HR, 4.66; 95% CI, 1.23-17.71; P = .0237). And in a separate model, HLA-B27 positivity was significantly associated with radiographic progression, conferring a nearly fourfold higher risk of progression (HR, 3.99; 95% CI, 1.10-14.49; P = .0353).
Asked if rheumatologists need to manage patients with nonradiographic axSpA differently than those with radiographic progression, Dr. Poddubnyy said that there was a small difference between the two in that biologics such as interleukin-17 inhibitors or Janus kinase inhibitors are approved for radiographic axSpA, whereas they are not approved for nonradiographic disease despite some off-label use. “We need to have high levels of symptoms plus nonresponse to NSAIDs and then we can prescribe a biologic,” he added.
For patients with nonradiographic axSpA, patients similarly need to have a high symptom burden and a nonresponse to NSAIDs, but in addition, physicians need to demonstrate objective signs of inflammatory activity, such as an elevated C-reactive protein level or the presence of inflammation on MRI before moving on the next level.
AbbVie funded the PROOF study. Dr. Poddubnyy declared receiving speaker bureau fees from AbbVie, Bristol-Myers Squibb, Eli Lilly, Merck Sharp & Dohme, Novartis, Pfizer, and UCB. He has also served as a consultant for AbbVie, Biocad, Eli Lilly, Gilead, GlaxoSmithKline, Merck Sharp & Dohme, Novartis, Pfizer, Samsung Bioepis, and UCB, as well as research support from AbbVie, Eli Lilly, Merck Sharp & Dohme, Novartis, and Pfizer. A number of coauthors also disclosed financial relationships with these and other pharmaceutical companies.
Four risk factors predicted the progression from nonradiographic to radiographic axial spondyloarthritis (axSpA) over a 5-year period in the PROOF study, a global, real-world, prospective, observational study carried out in 29 countries across six different geographic regions.
The predictors of progression within 5 years, based on the presence of radiographic sacroiliitis, included male gender, fulfillment of imaging criteria, HLA-B27 positivity, and a good response to NSAIDs, Denis Poddubnyy, MD, professor of rheumatology at Charité-Universitätsmedizin Berlin, said in his presentation of the study results at the annual European Congress of Rheumatology.
“In this study, 16% of nonradiographic axSpA patients progressed to radiographic axSpA within 5 years, with the mean time to disease progression of 2.4 years,” he said. PROOF (Patients with Axial Spondyloarthritis: Multicountry Registry of Clinical Characteristics) was originally designed to compare demographic and clinical characteristics of patients with axSpA across geographic regions.
In this particular analysis, Dr. Poddubnyy and colleagues aimed to track structural damage progression in the sacroiliac joint over time, as he explained in an interview. The study enrolled 2,633 adults with chronic back pain lasting for at least 3 months with onset before the age of 45 years. This analysis included patients diagnosed with axSpA who also fulfilled the Assessment of SpondyloArthritis international Society (ASAS) classification criteria for axSpA.
Both baseline and follow-up radiographs of sacroiliac joints were evaluated for those with an initial diagnosis of nonradiographic axSpA by two central readers; in cases when the readers disagreed on the classification – either nonradiographic or radiographic axSpA – images were adjudicated by a third reader. Radiographic progression from nonradiographic to radiographic axSpA was evaluated over the next 5 years.
Among all enrolled patients, 82% (n = 2,165) were diagnosed with axSpA and fulfilled the ASAS classification criteria. Of 1,612 who were classified by central reading, 65% had radiographic axSpA while the remaining 35% had nonradiographic axSpA. About 78% of those with nonradiographic axSpA fulfilled the ASAS classification criteria because of positive findings on imaging plus one or more features of spondyloarthritis. The other 22% were classified according to clinical criteria.
A total of 246 nonradiographic axSpA patients who had one or more follow-up radiographs of the sacroiliac joint were included in the current analysis. In this smaller group of patients, progression from the initial diagnosis of nonradiographic to radiographic axSpA at any one point over the 5-year follow-up occurred in 40 patients (16%) at a range of between 0.9 and 5.1 years.
“Females are more likely to stay in the nonradiographic stage than males,” Dr. Poddubnyy noted. Indeed, male gender conferred an over-threefold higher risk of radiographic progression, compared with females (hazard ratio, 3.16; 95% confidence interval, 1.22-8.17; P = .0174). Fulfillment of imaging criteria – in other words, the presence of inflammation on MRI – was also a strong predictor of progression, conferring an over-sixfold risk of radiographic progression (HR, 6.64; 95% CI, 1.37-32.25; P = .0188).
Interestingly, a good response to NSAIDs – the mainstay treatment for both nonradiographic and radiographic axSpA – was also significantly associated with radiographic progression, conferring an over-fourfold risk among those with an initial diagnosis of nonradiographic axSpA (HR, 4.66; 95% CI, 1.23-17.71; P = .0237). And in a separate model, HLA-B27 positivity was significantly associated with radiographic progression, conferring a nearly fourfold higher risk of progression (HR, 3.99; 95% CI, 1.10-14.49; P = .0353).
Asked if rheumatologists need to manage patients with nonradiographic axSpA differently than those with radiographic progression, Dr. Poddubnyy said that there was a small difference between the two in that biologics such as interleukin-17 inhibitors or Janus kinase inhibitors are approved for radiographic axSpA, whereas they are not approved for nonradiographic disease despite some off-label use. “We need to have high levels of symptoms plus nonresponse to NSAIDs and then we can prescribe a biologic,” he added.
For patients with nonradiographic axSpA, patients similarly need to have a high symptom burden and a nonresponse to NSAIDs, but in addition, physicians need to demonstrate objective signs of inflammatory activity, such as an elevated C-reactive protein level or the presence of inflammation on MRI before moving on the next level.
AbbVie funded the PROOF study. Dr. Poddubnyy declared receiving speaker bureau fees from AbbVie, Bristol-Myers Squibb, Eli Lilly, Merck Sharp & Dohme, Novartis, Pfizer, and UCB. He has also served as a consultant for AbbVie, Biocad, Eli Lilly, Gilead, GlaxoSmithKline, Merck Sharp & Dohme, Novartis, Pfizer, Samsung Bioepis, and UCB, as well as research support from AbbVie, Eli Lilly, Merck Sharp & Dohme, Novartis, and Pfizer. A number of coauthors also disclosed financial relationships with these and other pharmaceutical companies.
Four risk factors predicted the progression from nonradiographic to radiographic axial spondyloarthritis (axSpA) over a 5-year period in the PROOF study, a global, real-world, prospective, observational study carried out in 29 countries across six different geographic regions.
The predictors of progression within 5 years, based on the presence of radiographic sacroiliitis, included male gender, fulfillment of imaging criteria, HLA-B27 positivity, and a good response to NSAIDs, Denis Poddubnyy, MD, professor of rheumatology at Charité-Universitätsmedizin Berlin, said in his presentation of the study results at the annual European Congress of Rheumatology.
“In this study, 16% of nonradiographic axSpA patients progressed to radiographic axSpA within 5 years, with the mean time to disease progression of 2.4 years,” he said. PROOF (Patients with Axial Spondyloarthritis: Multicountry Registry of Clinical Characteristics) was originally designed to compare demographic and clinical characteristics of patients with axSpA across geographic regions.
In this particular analysis, Dr. Poddubnyy and colleagues aimed to track structural damage progression in the sacroiliac joint over time, as he explained in an interview. The study enrolled 2,633 adults with chronic back pain lasting for at least 3 months with onset before the age of 45 years. This analysis included patients diagnosed with axSpA who also fulfilled the Assessment of SpondyloArthritis international Society (ASAS) classification criteria for axSpA.
Both baseline and follow-up radiographs of sacroiliac joints were evaluated for those with an initial diagnosis of nonradiographic axSpA by two central readers; in cases when the readers disagreed on the classification – either nonradiographic or radiographic axSpA – images were adjudicated by a third reader. Radiographic progression from nonradiographic to radiographic axSpA was evaluated over the next 5 years.
Among all enrolled patients, 82% (n = 2,165) were diagnosed with axSpA and fulfilled the ASAS classification criteria. Of 1,612 who were classified by central reading, 65% had radiographic axSpA while the remaining 35% had nonradiographic axSpA. About 78% of those with nonradiographic axSpA fulfilled the ASAS classification criteria because of positive findings on imaging plus one or more features of spondyloarthritis. The other 22% were classified according to clinical criteria.
A total of 246 nonradiographic axSpA patients who had one or more follow-up radiographs of the sacroiliac joint were included in the current analysis. In this smaller group of patients, progression from the initial diagnosis of nonradiographic to radiographic axSpA at any one point over the 5-year follow-up occurred in 40 patients (16%) at a range of between 0.9 and 5.1 years.
“Females are more likely to stay in the nonradiographic stage than males,” Dr. Poddubnyy noted. Indeed, male gender conferred an over-threefold higher risk of radiographic progression, compared with females (hazard ratio, 3.16; 95% confidence interval, 1.22-8.17; P = .0174). Fulfillment of imaging criteria – in other words, the presence of inflammation on MRI – was also a strong predictor of progression, conferring an over-sixfold risk of radiographic progression (HR, 6.64; 95% CI, 1.37-32.25; P = .0188).
Interestingly, a good response to NSAIDs – the mainstay treatment for both nonradiographic and radiographic axSpA – was also significantly associated with radiographic progression, conferring an over-fourfold risk among those with an initial diagnosis of nonradiographic axSpA (HR, 4.66; 95% CI, 1.23-17.71; P = .0237). And in a separate model, HLA-B27 positivity was significantly associated with radiographic progression, conferring a nearly fourfold higher risk of progression (HR, 3.99; 95% CI, 1.10-14.49; P = .0353).
Asked if rheumatologists need to manage patients with nonradiographic axSpA differently than those with radiographic progression, Dr. Poddubnyy said that there was a small difference between the two in that biologics such as interleukin-17 inhibitors or Janus kinase inhibitors are approved for radiographic axSpA, whereas they are not approved for nonradiographic disease despite some off-label use. “We need to have high levels of symptoms plus nonresponse to NSAIDs and then we can prescribe a biologic,” he added.
For patients with nonradiographic axSpA, patients similarly need to have a high symptom burden and a nonresponse to NSAIDs, but in addition, physicians need to demonstrate objective signs of inflammatory activity, such as an elevated C-reactive protein level or the presence of inflammation on MRI before moving on the next level.
AbbVie funded the PROOF study. Dr. Poddubnyy declared receiving speaker bureau fees from AbbVie, Bristol-Myers Squibb, Eli Lilly, Merck Sharp & Dohme, Novartis, Pfizer, and UCB. He has also served as a consultant for AbbVie, Biocad, Eli Lilly, Gilead, GlaxoSmithKline, Merck Sharp & Dohme, Novartis, Pfizer, Samsung Bioepis, and UCB, as well as research support from AbbVie, Eli Lilly, Merck Sharp & Dohme, Novartis, and Pfizer. A number of coauthors also disclosed financial relationships with these and other pharmaceutical companies.
FROM THE EULAR 2022 CONGRESS
Acute exacerbations common and often fatal in RA-ILD
Acute exacerbations (AEs) are both common in rheumatoid arthritis–associated interstitial lung disease (RA-ILD) and are a frequent cause of imminent mortality, a retrospective Japanese study suggests.
The same is also true for patients with idiopathic pulmonary fibrosis (IPF) for whom an AE is the most frequent cause of death as well, the same comparative study indicates.
“Several studies have reported that acute exacerbation, which occurs during the clinical course of idiopathic pulmonary fibrosis (IPF), also occurs in rheumatoid arthritis–associated interstitial lung disease (RA-ILD),” lead author Junji Otsuka, MD, PhD, of the National Hospital Organization Omuta National Hospital, Fukuoka, Japan, and colleagues observed.
“[We found that] AE was not uncommon in RA-ILD or IPF ... but the prognosis after AE of RA-ILD was significantly better than that of IPF [even though] the most frequent cause of death in RA-ILD and IPF was AE,” they stated.
The study was published online in Respiratory Medicine.
Patient features
The study involved 149 RA-ILD patients with a median age of 72 years at RA onset. The median time from ILD diagnosis to onset of AE was 48.5 months, while the median survival time after the onset of AE was 196 days (range 1-3,463 days), as the authors detailed. “All patients were treated with corticosteroids,” the authors noted, and almost all of them were treated with steroid pulse therapy.
Noninvasive positive pressure ventilation (NPPV) was used to maintain oxygenation in 18.5% of patients with severe respiratory failure, while invasive positive pressure ventilation (IPPV) was used in almost 26% of patients with the same degree of respiratory failure. Features of patients who developed an AE were then compared with those who did not.
Interestingly, no significant differences in clinical parameters were seen between those who developed an AE and those who did not. Nor were there any significant differences between the 2 groups in the length of time from the ILD diagnosis to the development of an AE. Some 18% of RA-ILD patients developed an AE, as did over 27% of patients with IPF, investigators report.
, compared with only 60 days for those with IPF (P = .038). In a multivariable analysis, hypoalbuminemia at an odds ratio of .090 (95% confidence interval, 0.011-0.733; P = .012) as well as percent carbon monoxide diffusion capacity at an OR of .810 (95% CI, 0.814-0.964; P < .01) were both independent risk factors for the development of an AE, the investigators pointed out.
The best cut-off level for predicting the risk of an AE was 3.0 g/dL (95% CI, 0.011-0.733; P = .012) for serum albumin and 53% (95% CI, 0.814-0.964; P < .01) for carbon monoxide diffusion capacity. As Dr. Otsuka noted in an email to this news organization, low serum albumin likely correlates with a generally poor condition, while low carbon monoxide diffusion capacity is likely due to lung fibrosis.
“But if low albumin and low carbon monoxide diffusion capacity are due to the progression of ILD, both values may be difficult to improve,” he added.
Survivors versus nonsurvivors
Of those patients with RA-ILD who developed an AE, approximately half recovered. Among the IPF patients who developed an AE during the study period, approximately 39% recovered from the event, while 70% did not. Comparing RA-ILD patients who survived versus whose who did not, again, no significant demographic or clinical differences were seen between the 2 groups. On the other hand, the number of patients treated with immunosuppressants for their AE was significantly higher among patients who did not survive the AE, compared with those who did (P =.022), investigators note.
Similarly, the number of patients who required NPPV was also significantly higher among those who did not survive, compared with those who did. In fact, “none of the surviving patients used NPPV (P <0.01),” the authors stress. The number of patients who required IPPV was also significantly higher among nonsurvivors than among survivors (P =.017), and of the small number of patients who were treated with IPPV, all but one died without recovery.
As the authors suggested, these findings suggest that RA-ILD patients who recover from an AE with the help of corticosteroids alone have a relatively decent prognosis. In contrast, those who require immunosuppressive drugs in addition to steroids or mechanical ventilation for AE management can be expected to have a poor prognosis.
The same can also be said for IPF patients and even with the help of mechanical ventilation “with IPF patients, the survival rate is low anyway, so the indication for mechanical ventilation should be carefully judged,” Dr. Otsuka stressed.
Cause of death
The authors also compared the cause of death between patients with RA-ILD and those with IPF. “In RA-ILD patients, the most frequent cause of death was AE,” they report, at close to 35% of all patients with RA-ILD. This was also true for IPF patients among whom AE was the cause of death for over 44%. “These results indicate that, as in IPF, AE develops in the clinical course of RA-ILD with considerable frequency,” investigators note.
“During the clinical course of RA-ILD, as with IPD, it is necessary to pay attention to AE,” they stress. Dr. Otsuka added: “It may be difficult to change the prognosis of these patients.”
“However, knowing which patients are more likely to develop AE may help predict the prognosis, and it may be improved if antifibrotic agents are used for these patients,” he said. Elizabeth Volkmann, MD, director, UCLA scleroderma program, University of California, Los Angeles, felt that understanding the risk factors for AEs in this patient population may help physicians identify a subgroup of patients with RA-ILD who require closer monitoring and follow-up.
“These patients may also require more aggressive treatment for RA-ILD to prevent AEs,” she said in an email to this news organization. Given that the study was retrospective in nature, Dr. Volkmann cautioned that there were likely multiple confounding factors that could have affected survival in this patient population and not to take away from the study that survival was solely affected by immunosuppressant use, for example.
“It is possible that patients [treated with] immunosuppressants had other features of their disease that independently heightened their risk of mortality,” Dr. Volkmann said. Similarly, physicians should not assume that the high mortality rate seen in RA-ILD patients who were treated with mechanical ventilation had anything to do with mechanical ventilation itself, as patients requiring ventilation are likely to have worse outcomes, as she stressed.
As for hypoalbuminemia, Dr. Volkmann pointed out that hypoalbuminemia is often a sign of malnutrition in these patients. “Studies have demonstrated that malnutrition is an independent predictor of mortality in patients with ILD,” she emphasized.
“Optimizing patients’ nutritional status could potentially help lower the risk of AEs,” Dr. Volkmann suggested.
Limitations of the study include the fact that it was a single-center design study and included only a limited number of patients.
No specific funding source was noted. The authors have no conflicts of interest to declare.
Acute exacerbations (AEs) are both common in rheumatoid arthritis–associated interstitial lung disease (RA-ILD) and are a frequent cause of imminent mortality, a retrospective Japanese study suggests.
The same is also true for patients with idiopathic pulmonary fibrosis (IPF) for whom an AE is the most frequent cause of death as well, the same comparative study indicates.
“Several studies have reported that acute exacerbation, which occurs during the clinical course of idiopathic pulmonary fibrosis (IPF), also occurs in rheumatoid arthritis–associated interstitial lung disease (RA-ILD),” lead author Junji Otsuka, MD, PhD, of the National Hospital Organization Omuta National Hospital, Fukuoka, Japan, and colleagues observed.
“[We found that] AE was not uncommon in RA-ILD or IPF ... but the prognosis after AE of RA-ILD was significantly better than that of IPF [even though] the most frequent cause of death in RA-ILD and IPF was AE,” they stated.
The study was published online in Respiratory Medicine.
Patient features
The study involved 149 RA-ILD patients with a median age of 72 years at RA onset. The median time from ILD diagnosis to onset of AE was 48.5 months, while the median survival time after the onset of AE was 196 days (range 1-3,463 days), as the authors detailed. “All patients were treated with corticosteroids,” the authors noted, and almost all of them were treated with steroid pulse therapy.
Noninvasive positive pressure ventilation (NPPV) was used to maintain oxygenation in 18.5% of patients with severe respiratory failure, while invasive positive pressure ventilation (IPPV) was used in almost 26% of patients with the same degree of respiratory failure. Features of patients who developed an AE were then compared with those who did not.
Interestingly, no significant differences in clinical parameters were seen between those who developed an AE and those who did not. Nor were there any significant differences between the 2 groups in the length of time from the ILD diagnosis to the development of an AE. Some 18% of RA-ILD patients developed an AE, as did over 27% of patients with IPF, investigators report.
, compared with only 60 days for those with IPF (P = .038). In a multivariable analysis, hypoalbuminemia at an odds ratio of .090 (95% confidence interval, 0.011-0.733; P = .012) as well as percent carbon monoxide diffusion capacity at an OR of .810 (95% CI, 0.814-0.964; P < .01) were both independent risk factors for the development of an AE, the investigators pointed out.
The best cut-off level for predicting the risk of an AE was 3.0 g/dL (95% CI, 0.011-0.733; P = .012) for serum albumin and 53% (95% CI, 0.814-0.964; P < .01) for carbon monoxide diffusion capacity. As Dr. Otsuka noted in an email to this news organization, low serum albumin likely correlates with a generally poor condition, while low carbon monoxide diffusion capacity is likely due to lung fibrosis.
“But if low albumin and low carbon monoxide diffusion capacity are due to the progression of ILD, both values may be difficult to improve,” he added.
Survivors versus nonsurvivors
Of those patients with RA-ILD who developed an AE, approximately half recovered. Among the IPF patients who developed an AE during the study period, approximately 39% recovered from the event, while 70% did not. Comparing RA-ILD patients who survived versus whose who did not, again, no significant demographic or clinical differences were seen between the 2 groups. On the other hand, the number of patients treated with immunosuppressants for their AE was significantly higher among patients who did not survive the AE, compared with those who did (P =.022), investigators note.
Similarly, the number of patients who required NPPV was also significantly higher among those who did not survive, compared with those who did. In fact, “none of the surviving patients used NPPV (P <0.01),” the authors stress. The number of patients who required IPPV was also significantly higher among nonsurvivors than among survivors (P =.017), and of the small number of patients who were treated with IPPV, all but one died without recovery.
As the authors suggested, these findings suggest that RA-ILD patients who recover from an AE with the help of corticosteroids alone have a relatively decent prognosis. In contrast, those who require immunosuppressive drugs in addition to steroids or mechanical ventilation for AE management can be expected to have a poor prognosis.
The same can also be said for IPF patients and even with the help of mechanical ventilation “with IPF patients, the survival rate is low anyway, so the indication for mechanical ventilation should be carefully judged,” Dr. Otsuka stressed.
Cause of death
The authors also compared the cause of death between patients with RA-ILD and those with IPF. “In RA-ILD patients, the most frequent cause of death was AE,” they report, at close to 35% of all patients with RA-ILD. This was also true for IPF patients among whom AE was the cause of death for over 44%. “These results indicate that, as in IPF, AE develops in the clinical course of RA-ILD with considerable frequency,” investigators note.
“During the clinical course of RA-ILD, as with IPD, it is necessary to pay attention to AE,” they stress. Dr. Otsuka added: “It may be difficult to change the prognosis of these patients.”
“However, knowing which patients are more likely to develop AE may help predict the prognosis, and it may be improved if antifibrotic agents are used for these patients,” he said. Elizabeth Volkmann, MD, director, UCLA scleroderma program, University of California, Los Angeles, felt that understanding the risk factors for AEs in this patient population may help physicians identify a subgroup of patients with RA-ILD who require closer monitoring and follow-up.
“These patients may also require more aggressive treatment for RA-ILD to prevent AEs,” she said in an email to this news organization. Given that the study was retrospective in nature, Dr. Volkmann cautioned that there were likely multiple confounding factors that could have affected survival in this patient population and not to take away from the study that survival was solely affected by immunosuppressant use, for example.
“It is possible that patients [treated with] immunosuppressants had other features of their disease that independently heightened their risk of mortality,” Dr. Volkmann said. Similarly, physicians should not assume that the high mortality rate seen in RA-ILD patients who were treated with mechanical ventilation had anything to do with mechanical ventilation itself, as patients requiring ventilation are likely to have worse outcomes, as she stressed.
As for hypoalbuminemia, Dr. Volkmann pointed out that hypoalbuminemia is often a sign of malnutrition in these patients. “Studies have demonstrated that malnutrition is an independent predictor of mortality in patients with ILD,” she emphasized.
“Optimizing patients’ nutritional status could potentially help lower the risk of AEs,” Dr. Volkmann suggested.
Limitations of the study include the fact that it was a single-center design study and included only a limited number of patients.
No specific funding source was noted. The authors have no conflicts of interest to declare.
Acute exacerbations (AEs) are both common in rheumatoid arthritis–associated interstitial lung disease (RA-ILD) and are a frequent cause of imminent mortality, a retrospective Japanese study suggests.
The same is also true for patients with idiopathic pulmonary fibrosis (IPF) for whom an AE is the most frequent cause of death as well, the same comparative study indicates.
“Several studies have reported that acute exacerbation, which occurs during the clinical course of idiopathic pulmonary fibrosis (IPF), also occurs in rheumatoid arthritis–associated interstitial lung disease (RA-ILD),” lead author Junji Otsuka, MD, PhD, of the National Hospital Organization Omuta National Hospital, Fukuoka, Japan, and colleagues observed.
“[We found that] AE was not uncommon in RA-ILD or IPF ... but the prognosis after AE of RA-ILD was significantly better than that of IPF [even though] the most frequent cause of death in RA-ILD and IPF was AE,” they stated.
The study was published online in Respiratory Medicine.
Patient features
The study involved 149 RA-ILD patients with a median age of 72 years at RA onset. The median time from ILD diagnosis to onset of AE was 48.5 months, while the median survival time after the onset of AE was 196 days (range 1-3,463 days), as the authors detailed. “All patients were treated with corticosteroids,” the authors noted, and almost all of them were treated with steroid pulse therapy.
Noninvasive positive pressure ventilation (NPPV) was used to maintain oxygenation in 18.5% of patients with severe respiratory failure, while invasive positive pressure ventilation (IPPV) was used in almost 26% of patients with the same degree of respiratory failure. Features of patients who developed an AE were then compared with those who did not.
Interestingly, no significant differences in clinical parameters were seen between those who developed an AE and those who did not. Nor were there any significant differences between the 2 groups in the length of time from the ILD diagnosis to the development of an AE. Some 18% of RA-ILD patients developed an AE, as did over 27% of patients with IPF, investigators report.
, compared with only 60 days for those with IPF (P = .038). In a multivariable analysis, hypoalbuminemia at an odds ratio of .090 (95% confidence interval, 0.011-0.733; P = .012) as well as percent carbon monoxide diffusion capacity at an OR of .810 (95% CI, 0.814-0.964; P < .01) were both independent risk factors for the development of an AE, the investigators pointed out.
The best cut-off level for predicting the risk of an AE was 3.0 g/dL (95% CI, 0.011-0.733; P = .012) for serum albumin and 53% (95% CI, 0.814-0.964; P < .01) for carbon monoxide diffusion capacity. As Dr. Otsuka noted in an email to this news organization, low serum albumin likely correlates with a generally poor condition, while low carbon monoxide diffusion capacity is likely due to lung fibrosis.
“But if low albumin and low carbon monoxide diffusion capacity are due to the progression of ILD, both values may be difficult to improve,” he added.
Survivors versus nonsurvivors
Of those patients with RA-ILD who developed an AE, approximately half recovered. Among the IPF patients who developed an AE during the study period, approximately 39% recovered from the event, while 70% did not. Comparing RA-ILD patients who survived versus whose who did not, again, no significant demographic or clinical differences were seen between the 2 groups. On the other hand, the number of patients treated with immunosuppressants for their AE was significantly higher among patients who did not survive the AE, compared with those who did (P =.022), investigators note.
Similarly, the number of patients who required NPPV was also significantly higher among those who did not survive, compared with those who did. In fact, “none of the surviving patients used NPPV (P <0.01),” the authors stress. The number of patients who required IPPV was also significantly higher among nonsurvivors than among survivors (P =.017), and of the small number of patients who were treated with IPPV, all but one died without recovery.
As the authors suggested, these findings suggest that RA-ILD patients who recover from an AE with the help of corticosteroids alone have a relatively decent prognosis. In contrast, those who require immunosuppressive drugs in addition to steroids or mechanical ventilation for AE management can be expected to have a poor prognosis.
The same can also be said for IPF patients and even with the help of mechanical ventilation “with IPF patients, the survival rate is low anyway, so the indication for mechanical ventilation should be carefully judged,” Dr. Otsuka stressed.
Cause of death
The authors also compared the cause of death between patients with RA-ILD and those with IPF. “In RA-ILD patients, the most frequent cause of death was AE,” they report, at close to 35% of all patients with RA-ILD. This was also true for IPF patients among whom AE was the cause of death for over 44%. “These results indicate that, as in IPF, AE develops in the clinical course of RA-ILD with considerable frequency,” investigators note.
“During the clinical course of RA-ILD, as with IPD, it is necessary to pay attention to AE,” they stress. Dr. Otsuka added: “It may be difficult to change the prognosis of these patients.”
“However, knowing which patients are more likely to develop AE may help predict the prognosis, and it may be improved if antifibrotic agents are used for these patients,” he said. Elizabeth Volkmann, MD, director, UCLA scleroderma program, University of California, Los Angeles, felt that understanding the risk factors for AEs in this patient population may help physicians identify a subgroup of patients with RA-ILD who require closer monitoring and follow-up.
“These patients may also require more aggressive treatment for RA-ILD to prevent AEs,” she said in an email to this news organization. Given that the study was retrospective in nature, Dr. Volkmann cautioned that there were likely multiple confounding factors that could have affected survival in this patient population and not to take away from the study that survival was solely affected by immunosuppressant use, for example.
“It is possible that patients [treated with] immunosuppressants had other features of their disease that independently heightened their risk of mortality,” Dr. Volkmann said. Similarly, physicians should not assume that the high mortality rate seen in RA-ILD patients who were treated with mechanical ventilation had anything to do with mechanical ventilation itself, as patients requiring ventilation are likely to have worse outcomes, as she stressed.
As for hypoalbuminemia, Dr. Volkmann pointed out that hypoalbuminemia is often a sign of malnutrition in these patients. “Studies have demonstrated that malnutrition is an independent predictor of mortality in patients with ILD,” she emphasized.
“Optimizing patients’ nutritional status could potentially help lower the risk of AEs,” Dr. Volkmann suggested.
Limitations of the study include the fact that it was a single-center design study and included only a limited number of patients.
No specific funding source was noted. The authors have no conflicts of interest to declare.