FL: Tafasitamab Plus Combo Boosts Outcomes

Article Type
Changed
Wed, 12/18/2024 - 13:23

In the treatment of relapsed or refractory follicular lymphoma (R/R FL), the addition of tafasitamab to a standard 2-drug combination of lenalidomide and rituximab (len+R) significantly improves progression-free survival, with no increases in safety events.

“This study is the first to validate combining two monoclonal antibodies (anti-CD19 with anti-CD20) in the treatment of lymphoma,” said first author Laurie H. Sehn, MD, MPH, a clinical professor with the BC Cancer Centre for Lymphoid Cancer and the University of British Columbia in Vancouver, Canada. She presented the findings at the American Society of Hematology 2024 Annual Meeting.

“Tafasitamab plus lenalidomide and rituximab can be administered in community as well as academic settings and represents a potential new standard-of-care option for patients with R/R follicular lymphoma,” she noted.

The results are from the international, phase 3 inMIND multicenter trial involving 548 patients with R/R FL, with a median age of 64. Of the patients, 45% had been treated with two or more prior lines of therapy.

At a median follow up of 14.1 months, those randomized to treatment with tafasitamab and len+R (n = 273) had a significantly lower risk for progression, relapse, or death than those receiving the double therapy with placebo (n = 275), with a median progression-free survival of 22.4 months vs 13.9 months, respectively (hazard ratio [HR], 0.43; P < .0001).

Improved progression-free survival was observed across all prespecified subgroups, including patients with disease progression within 24 months, those who were refractory to prior anti-CD20 monoclonal antibodies, and who received multiple prior lines of therapy.

“Although this study was not powered for individual subgroups, it is clear that there’s a significant benefit of tafasitamab in all subgroups regardless of status of [disease progression within 24 months] and regardless of refractoriness to prior anti-CD20 monoclonal antibodies,” Sehn noted.

FL, the most common, indolent form of B-cell non-Hodgkin lymphoma (NHL), is commonly treated with frontline therapy of chemoimmunotherapy; however, response durations begin to dwindle after successive lines of treatment.

Lenalidomide and rituximab are approved and commonly used in the treatment of FL after more than one prior line of treatment.

With tafasitamab, which is administered intravenously, already having been approved for use in combination with lenalidomide in the treatment of R/R diffuse large B-cell lymphoma, based on results of the previous L-MIND study, Sehn and colleagues investigated its benefits in FL or marginal zone NHL.

Of the patients included in the trial, 55% were men and 79% had intermediate or high-risk FLIPI scores, referring to the FL International Prognostic Index, a scoring system used for predicting prognoses of patients with FL.

Treatment in the study consisted of 12 mg/kg intravenous tafasitamab or placebo on days 1, 8, 15, and 22 of cycles 1-3 and days 1 and 15 of cycles 4-12, with standard dosing of len+R for up to 12 cycles of 28 days each. 

In terms of the data cutoff, patients in the tafasitamab arm received a median of 12 cycles of treatment vs 11 cycles for placebo, and 19% and 15% were still on treatment, respectively, at the cut-off. 

Of the patients, 81% and 84%, respectively, had discontinued treatment primarily because of treatment completion (54% and 43%) or disease progression (11% and 31%). 

In addition to the progression-free survival primary endpoint, the tafasitamab arm also had a higher rate of complete response (CR) on PET (49.4% vs 39.8%; P = .029) and higher overall response rate (83.5% vs 72.4%; P = .0014) than the placebo arm. 

The duration of response was also higher with tafasitamab (median, 21.2 months vs 13.6 months; HR, 0.47; P < .0001).

Overall survival data, though immature, also favored tafasitamab (HR, 0.59). 

The two arms had similar rates of treatment-emergent adverse events, with similar toxicity profiles and no new significant safety signals related to tafasitamab. The rates of discontinuations and dose reductions were similar, with a median dose intensity of 86% with tafasitamab vs 87%.

The most common grade 3 or 4 adverse events were similar between the tafasitamab and placebo groups, including neutropenia (40% vs 38%, respectively), pneumonia (8% vs 5%), thrombocytopenia (6% vs 7%), decreased neutrophils (6% vs 7%), COVID-19 (6% vs 2%), and COVID-19 pneumonia (5% vs 1%). 

“Importantly, the addition of tafasitamab did not impede the delivery of lenalidomide and rituximab, with similar observed dose discontinuations or interruptions in both cohorts,” Sehn said.

“The inMIND phase 3 study met its primary endpoint of improved progression-free survival with the addition of tafasitamab to lenalidomide and rituximab in patients with R/R follicular lymphoma, representing a 57% reduction in the risk of progression, relapse, or death,” Sehn said.

Commenting on the study, Juan Pablo Alderuccio, MD, an associate professor of medicine, Division of Hematology in the Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine in Florida, said the findings are notable in that “this is the first time two monoclonal antibodies have been tested in follicular lymphoma.”

“The study demonstrates that simultaneously targeting CD19 and CD20 improves outcomes,” he said in an interview.

Alderuccio noted the key caveats include that “PET/CT complete response correlates well with survival in follicular lymphoma. In this study, the PET/CT CR rate was 49.4%, underscoring the need for longer follow-ups to better assess those responses’ durability.”

“Another caveat of this regimen is the treatment schedule, which requires weekly tafasitamab infusions during cycles 1-3 and every 2 weeks during cycles 4-12. This is associated with rituximab and lenalidomide administration,” he said.

Ultimately, “the results underscore the potential of tafasitamab in combination with lenalidomide and rituximab to become a new treatment option in the second-line or later follicular lymphoma.”

“However, I would like to see more follow-up data before considering it a new standard of care,” he cautioned.

The study was funded by Incyte. Sehn reported ties with AbbVie, Amgen, AstraZeneca, BeiGene, BMS/Celgene, Genmab, Kite/Gilead, Incyte, Janssen, and Merck. Alderuccio disclosed relationships with Genmab, ADC Therapeutics, BeiGene, AbbVie, Genentech, Novartis, Regeneron, and Lilly.

A version of this article appeared on Medscape.com.

Meeting/Event
Publications
Topics
Sections
Meeting/Event
Meeting/Event

In the treatment of relapsed or refractory follicular lymphoma (R/R FL), the addition of tafasitamab to a standard 2-drug combination of lenalidomide and rituximab (len+R) significantly improves progression-free survival, with no increases in safety events.

“This study is the first to validate combining two monoclonal antibodies (anti-CD19 with anti-CD20) in the treatment of lymphoma,” said first author Laurie H. Sehn, MD, MPH, a clinical professor with the BC Cancer Centre for Lymphoid Cancer and the University of British Columbia in Vancouver, Canada. She presented the findings at the American Society of Hematology 2024 Annual Meeting.

“Tafasitamab plus lenalidomide and rituximab can be administered in community as well as academic settings and represents a potential new standard-of-care option for patients with R/R follicular lymphoma,” she noted.

The results are from the international, phase 3 inMIND multicenter trial involving 548 patients with R/R FL, with a median age of 64. Of the patients, 45% had been treated with two or more prior lines of therapy.

At a median follow up of 14.1 months, those randomized to treatment with tafasitamab and len+R (n = 273) had a significantly lower risk for progression, relapse, or death than those receiving the double therapy with placebo (n = 275), with a median progression-free survival of 22.4 months vs 13.9 months, respectively (hazard ratio [HR], 0.43; P < .0001).

Improved progression-free survival was observed across all prespecified subgroups, including patients with disease progression within 24 months, those who were refractory to prior anti-CD20 monoclonal antibodies, and who received multiple prior lines of therapy.

“Although this study was not powered for individual subgroups, it is clear that there’s a significant benefit of tafasitamab in all subgroups regardless of status of [disease progression within 24 months] and regardless of refractoriness to prior anti-CD20 monoclonal antibodies,” Sehn noted.

FL, the most common, indolent form of B-cell non-Hodgkin lymphoma (NHL), is commonly treated with frontline therapy of chemoimmunotherapy; however, response durations begin to dwindle after successive lines of treatment.

Lenalidomide and rituximab are approved and commonly used in the treatment of FL after more than one prior line of treatment.

With tafasitamab, which is administered intravenously, already having been approved for use in combination with lenalidomide in the treatment of R/R diffuse large B-cell lymphoma, based on results of the previous L-MIND study, Sehn and colleagues investigated its benefits in FL or marginal zone NHL.

Of the patients included in the trial, 55% were men and 79% had intermediate or high-risk FLIPI scores, referring to the FL International Prognostic Index, a scoring system used for predicting prognoses of patients with FL.

Treatment in the study consisted of 12 mg/kg intravenous tafasitamab or placebo on days 1, 8, 15, and 22 of cycles 1-3 and days 1 and 15 of cycles 4-12, with standard dosing of len+R for up to 12 cycles of 28 days each. 

In terms of the data cutoff, patients in the tafasitamab arm received a median of 12 cycles of treatment vs 11 cycles for placebo, and 19% and 15% were still on treatment, respectively, at the cut-off. 

Of the patients, 81% and 84%, respectively, had discontinued treatment primarily because of treatment completion (54% and 43%) or disease progression (11% and 31%). 

In addition to the progression-free survival primary endpoint, the tafasitamab arm also had a higher rate of complete response (CR) on PET (49.4% vs 39.8%; P = .029) and higher overall response rate (83.5% vs 72.4%; P = .0014) than the placebo arm. 

The duration of response was also higher with tafasitamab (median, 21.2 months vs 13.6 months; HR, 0.47; P < .0001).

Overall survival data, though immature, also favored tafasitamab (HR, 0.59). 

The two arms had similar rates of treatment-emergent adverse events, with similar toxicity profiles and no new significant safety signals related to tafasitamab. The rates of discontinuations and dose reductions were similar, with a median dose intensity of 86% with tafasitamab vs 87%.

The most common grade 3 or 4 adverse events were similar between the tafasitamab and placebo groups, including neutropenia (40% vs 38%, respectively), pneumonia (8% vs 5%), thrombocytopenia (6% vs 7%), decreased neutrophils (6% vs 7%), COVID-19 (6% vs 2%), and COVID-19 pneumonia (5% vs 1%). 

“Importantly, the addition of tafasitamab did not impede the delivery of lenalidomide and rituximab, with similar observed dose discontinuations or interruptions in both cohorts,” Sehn said.

“The inMIND phase 3 study met its primary endpoint of improved progression-free survival with the addition of tafasitamab to lenalidomide and rituximab in patients with R/R follicular lymphoma, representing a 57% reduction in the risk of progression, relapse, or death,” Sehn said.

Commenting on the study, Juan Pablo Alderuccio, MD, an associate professor of medicine, Division of Hematology in the Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine in Florida, said the findings are notable in that “this is the first time two monoclonal antibodies have been tested in follicular lymphoma.”

“The study demonstrates that simultaneously targeting CD19 and CD20 improves outcomes,” he said in an interview.

Alderuccio noted the key caveats include that “PET/CT complete response correlates well with survival in follicular lymphoma. In this study, the PET/CT CR rate was 49.4%, underscoring the need for longer follow-ups to better assess those responses’ durability.”

“Another caveat of this regimen is the treatment schedule, which requires weekly tafasitamab infusions during cycles 1-3 and every 2 weeks during cycles 4-12. This is associated with rituximab and lenalidomide administration,” he said.

Ultimately, “the results underscore the potential of tafasitamab in combination with lenalidomide and rituximab to become a new treatment option in the second-line or later follicular lymphoma.”

“However, I would like to see more follow-up data before considering it a new standard of care,” he cautioned.

The study was funded by Incyte. Sehn reported ties with AbbVie, Amgen, AstraZeneca, BeiGene, BMS/Celgene, Genmab, Kite/Gilead, Incyte, Janssen, and Merck. Alderuccio disclosed relationships with Genmab, ADC Therapeutics, BeiGene, AbbVie, Genentech, Novartis, Regeneron, and Lilly.

A version of this article appeared on Medscape.com.

In the treatment of relapsed or refractory follicular lymphoma (R/R FL), the addition of tafasitamab to a standard 2-drug combination of lenalidomide and rituximab (len+R) significantly improves progression-free survival, with no increases in safety events.

“This study is the first to validate combining two monoclonal antibodies (anti-CD19 with anti-CD20) in the treatment of lymphoma,” said first author Laurie H. Sehn, MD, MPH, a clinical professor with the BC Cancer Centre for Lymphoid Cancer and the University of British Columbia in Vancouver, Canada. She presented the findings at the American Society of Hematology 2024 Annual Meeting.

“Tafasitamab plus lenalidomide and rituximab can be administered in community as well as academic settings and represents a potential new standard-of-care option for patients with R/R follicular lymphoma,” she noted.

The results are from the international, phase 3 inMIND multicenter trial involving 548 patients with R/R FL, with a median age of 64. Of the patients, 45% had been treated with two or more prior lines of therapy.

At a median follow up of 14.1 months, those randomized to treatment with tafasitamab and len+R (n = 273) had a significantly lower risk for progression, relapse, or death than those receiving the double therapy with placebo (n = 275), with a median progression-free survival of 22.4 months vs 13.9 months, respectively (hazard ratio [HR], 0.43; P < .0001).

Improved progression-free survival was observed across all prespecified subgroups, including patients with disease progression within 24 months, those who were refractory to prior anti-CD20 monoclonal antibodies, and who received multiple prior lines of therapy.

“Although this study was not powered for individual subgroups, it is clear that there’s a significant benefit of tafasitamab in all subgroups regardless of status of [disease progression within 24 months] and regardless of refractoriness to prior anti-CD20 monoclonal antibodies,” Sehn noted.

FL, the most common, indolent form of B-cell non-Hodgkin lymphoma (NHL), is commonly treated with frontline therapy of chemoimmunotherapy; however, response durations begin to dwindle after successive lines of treatment.

Lenalidomide and rituximab are approved and commonly used in the treatment of FL after more than one prior line of treatment.

With tafasitamab, which is administered intravenously, already having been approved for use in combination with lenalidomide in the treatment of R/R diffuse large B-cell lymphoma, based on results of the previous L-MIND study, Sehn and colleagues investigated its benefits in FL or marginal zone NHL.

Of the patients included in the trial, 55% were men and 79% had intermediate or high-risk FLIPI scores, referring to the FL International Prognostic Index, a scoring system used for predicting prognoses of patients with FL.

Treatment in the study consisted of 12 mg/kg intravenous tafasitamab or placebo on days 1, 8, 15, and 22 of cycles 1-3 and days 1 and 15 of cycles 4-12, with standard dosing of len+R for up to 12 cycles of 28 days each. 

In terms of the data cutoff, patients in the tafasitamab arm received a median of 12 cycles of treatment vs 11 cycles for placebo, and 19% and 15% were still on treatment, respectively, at the cut-off. 

Of the patients, 81% and 84%, respectively, had discontinued treatment primarily because of treatment completion (54% and 43%) or disease progression (11% and 31%). 

In addition to the progression-free survival primary endpoint, the tafasitamab arm also had a higher rate of complete response (CR) on PET (49.4% vs 39.8%; P = .029) and higher overall response rate (83.5% vs 72.4%; P = .0014) than the placebo arm. 

The duration of response was also higher with tafasitamab (median, 21.2 months vs 13.6 months; HR, 0.47; P < .0001).

Overall survival data, though immature, also favored tafasitamab (HR, 0.59). 

The two arms had similar rates of treatment-emergent adverse events, with similar toxicity profiles and no new significant safety signals related to tafasitamab. The rates of discontinuations and dose reductions were similar, with a median dose intensity of 86% with tafasitamab vs 87%.

The most common grade 3 or 4 adverse events were similar between the tafasitamab and placebo groups, including neutropenia (40% vs 38%, respectively), pneumonia (8% vs 5%), thrombocytopenia (6% vs 7%), decreased neutrophils (6% vs 7%), COVID-19 (6% vs 2%), and COVID-19 pneumonia (5% vs 1%). 

“Importantly, the addition of tafasitamab did not impede the delivery of lenalidomide and rituximab, with similar observed dose discontinuations or interruptions in both cohorts,” Sehn said.

“The inMIND phase 3 study met its primary endpoint of improved progression-free survival with the addition of tafasitamab to lenalidomide and rituximab in patients with R/R follicular lymphoma, representing a 57% reduction in the risk of progression, relapse, or death,” Sehn said.

Commenting on the study, Juan Pablo Alderuccio, MD, an associate professor of medicine, Division of Hematology in the Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine in Florida, said the findings are notable in that “this is the first time two monoclonal antibodies have been tested in follicular lymphoma.”

“The study demonstrates that simultaneously targeting CD19 and CD20 improves outcomes,” he said in an interview.

Alderuccio noted the key caveats include that “PET/CT complete response correlates well with survival in follicular lymphoma. In this study, the PET/CT CR rate was 49.4%, underscoring the need for longer follow-ups to better assess those responses’ durability.”

“Another caveat of this regimen is the treatment schedule, which requires weekly tafasitamab infusions during cycles 1-3 and every 2 weeks during cycles 4-12. This is associated with rituximab and lenalidomide administration,” he said.

Ultimately, “the results underscore the potential of tafasitamab in combination with lenalidomide and rituximab to become a new treatment option in the second-line or later follicular lymphoma.”

“However, I would like to see more follow-up data before considering it a new standard of care,” he cautioned.

The study was funded by Incyte. Sehn reported ties with AbbVie, Amgen, AstraZeneca, BeiGene, BMS/Celgene, Genmab, Kite/Gilead, Incyte, Janssen, and Merck. Alderuccio disclosed relationships with Genmab, ADC Therapeutics, BeiGene, AbbVie, Genentech, Novartis, Regeneron, and Lilly.

A version of this article appeared on Medscape.com.

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM ASH 2024

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Wed, 12/18/2024 - 13:22
Un-Gate On Date
Wed, 12/18/2024 - 13:22
Use ProPublica
CFC Schedule Remove Status
Wed, 12/18/2024 - 13:22
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
survey writer start date
Wed, 12/18/2024 - 13:22

Angiotensin Receptor Blockers Cut Epilepsy Risk in Patients With Hypertension

Article Type
Changed
Tue, 12/17/2024 - 16:14

Angiotensin receptor blockers are more effective than other antihypertensive medications in reducing the risk for post-stroke epilepsy (PSE), new research showed.

Investigators found angiotensin receptor blockers are better at cutting the PSE risk in individuals with high blood pressure than angiotensin-converting enzyme (ACE) inhibitors, calcium channel blockers, and beta-blockers.

The results suggested angiotensin receptor blockers could be a frontline strategy to proactively forestall development of epilepsy in patients with a history of stroke and hypertension, said the study’s co-lead investigator Giacomo Evangelista, MD, PhD, a resident in neurology at the Epilepsy Center at “G. d’Annunzio” University of Chieti-Pescara, Italy.

The findings were presented at the American Epilepsy Society (AES) 78th Annual Meeting 2024.

 

Stroke and Seizures Tightly Linked

Stroke is the most common cause of seizures in patients older than 60 years. About 6%-8% of new epilepsy diagnoses in older adult patients are associated with a brain ischemic event.

Hypertension is among the most common risk factors for both epilepsy and stroke. The European Society of Cardiology recommends ACE inhibitors and angiotensin receptor blockers be considered first-line hypertension treatments.

Angiotensin receptor blockers seem to provide a protective effect in terms of seizures in the general population, but their possible preventive role in PSE has not been clear.

The retrospective, observational study included 528 patients (mean age, 71.4 years; 57.19% men) with hypertension and ischemic stroke without a history of epilepsy.

Researchers divided patients into those who developed PSE during the 2-year follow-up and those who didn’t. The main outcome was incidence of PSE associated with angiotensin receptor blocker therapy compared with other antihypertensive drug classes (ACE inhibitors, calcium channel blockers, and beta blockers).

Of the total, 7.2% of patients developed PSE. The study found patients treated with an angiotensin receptor blockers had a lower risk for PSE (P = .009). PSE incidence was higher among patients receiving calcium channel blockers (P = .019) and beta blockers (P = .008), but ACE inhibitors did not appear to affect PSE risk.

Further analysis showed that patients taking a beta blocker were 120% more likely to develop PSE, and those taking a calcium channel blocker were 110% more likely to develop PSE than those taking an angiotensin receptor blocker. The corresponding comparison was 65% for those taking an ACE inhibitor.

 

Potential Mechanisms

Angiotensin receptor blockers block the angiotensin II type 1 receptor, which may lead to numerous neuroprotective benefits such as improved blood flow to the brain and less neuroinflammation. The blockage may also reduce epilepsy severity and seizure frequency.

ACE inhibitors work in the same system as angiotensin receptor blockers but don’t bind directly to the angiotensin receptor. Experts believe these drugs may lead to inflammation that raises the risk for PSE.

Calcium channel blockers and beta blockers may increase the likelihood of seizures by inducing excessive excitability in the brain, which can trigger seizures.

Other research presented at the meeting found that angiotensin receptor blockers reduce the risk for epilepsy in patients with hypertension even in the absence of stroke.

Using data from an US national administrative database, the retrospective cohort study included 2,261,964 eligible adult beneficiaries diagnosed with hypertension and dispensed at least one angiotensin receptor blocker, ACE inhibitor, beta blocker, or calcium channel blocker from 2010 to 2017.

The study found angiotensin receptor blockers were associated with lower epilepsy incidence than ACE inhibitors (hazard ratio [HR], 0.75; 95% CI, 0.58-0.96), beta blockers (HR, 0.70; 95% CI, 0.54-0.90), and calcium channel blockers (HR, 0.80; 95% CI, 0.61-1.04).

The effect was most robust for the angiotensin receptor blocker losartan and among patients with no preexisting stroke or cardiovascular conditions.

Researchers ran the analysis excluding stroke and the association was still there, said lead study author Kimford Meador, MD, professor of neurology and neurological sciences, Stanford University, Palo Alto, California. “People with hypertension who use angiotensin receptor blockers — even if they don’t have a stroke — are less likely to develop epilepsy than if they use other major anti-hypertensives.”

 

Exciting and Suggestive

These new studies are “exciting” and “very suggestive,” said Meador. “We don’t have any true antiepileptic drugs; we have antiseizure medications. If we could reduce the incidence of epilepsy, that would be a very big thing.”

But the research to date hasn’t been “definitive” and is subject to bias, he said. “What we really need is a large randomized controlled trial to see if this is real because any observational study in humans has the potential for confounding from unmeasured variables.”

If such a study does show the effect is real, “that would change clinical practice,” said Meador.

As angiotensin receptor blockers have anti-inflammatory effects, researchers are now investigating whether this class of drugs might be useful in other diseases linked to inflammation such as multiple sclerosis, said Meador.

Commenting on the research, Alain Zingraff Looti, MD, assistant professor of neurology and public health sciences, Penn State College of Medicine, Hershey, Pennsylvania, said this new research is “important” although still at a “very preliminary stage.”

As the studies so far have all been observational, “they showed an association, but that doesn’t mean causality,” said Looti. These studies just looked at known potential confounders, “and there may be some unknown confounders that could explain the association,” he added.

But he acknowledged the results make sense from a biological standpoint. He noted that infusing angiotensin receptor blockers into rats causes a “cascade reaction” leading to reduced inflammation in the brain and then to less epilepsy. “Inflammation plays a major role in several models of epilepsy,” he added.

While these new results are important, Looti, too, would like to see a randomized controlled trial to confirm the findings.

Looti and Meador reported no relevant financial disclosures.

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

Meeting/Event
Publications
Topics
Sections
Meeting/Event
Meeting/Event

Angiotensin receptor blockers are more effective than other antihypertensive medications in reducing the risk for post-stroke epilepsy (PSE), new research showed.

Investigators found angiotensin receptor blockers are better at cutting the PSE risk in individuals with high blood pressure than angiotensin-converting enzyme (ACE) inhibitors, calcium channel blockers, and beta-blockers.

The results suggested angiotensin receptor blockers could be a frontline strategy to proactively forestall development of epilepsy in patients with a history of stroke and hypertension, said the study’s co-lead investigator Giacomo Evangelista, MD, PhD, a resident in neurology at the Epilepsy Center at “G. d’Annunzio” University of Chieti-Pescara, Italy.

The findings were presented at the American Epilepsy Society (AES) 78th Annual Meeting 2024.

 

Stroke and Seizures Tightly Linked

Stroke is the most common cause of seizures in patients older than 60 years. About 6%-8% of new epilepsy diagnoses in older adult patients are associated with a brain ischemic event.

Hypertension is among the most common risk factors for both epilepsy and stroke. The European Society of Cardiology recommends ACE inhibitors and angiotensin receptor blockers be considered first-line hypertension treatments.

Angiotensin receptor blockers seem to provide a protective effect in terms of seizures in the general population, but their possible preventive role in PSE has not been clear.

The retrospective, observational study included 528 patients (mean age, 71.4 years; 57.19% men) with hypertension and ischemic stroke without a history of epilepsy.

Researchers divided patients into those who developed PSE during the 2-year follow-up and those who didn’t. The main outcome was incidence of PSE associated with angiotensin receptor blocker therapy compared with other antihypertensive drug classes (ACE inhibitors, calcium channel blockers, and beta blockers).

Of the total, 7.2% of patients developed PSE. The study found patients treated with an angiotensin receptor blockers had a lower risk for PSE (P = .009). PSE incidence was higher among patients receiving calcium channel blockers (P = .019) and beta blockers (P = .008), but ACE inhibitors did not appear to affect PSE risk.

Further analysis showed that patients taking a beta blocker were 120% more likely to develop PSE, and those taking a calcium channel blocker were 110% more likely to develop PSE than those taking an angiotensin receptor blocker. The corresponding comparison was 65% for those taking an ACE inhibitor.

 

Potential Mechanisms

Angiotensin receptor blockers block the angiotensin II type 1 receptor, which may lead to numerous neuroprotective benefits such as improved blood flow to the brain and less neuroinflammation. The blockage may also reduce epilepsy severity and seizure frequency.

ACE inhibitors work in the same system as angiotensin receptor blockers but don’t bind directly to the angiotensin receptor. Experts believe these drugs may lead to inflammation that raises the risk for PSE.

Calcium channel blockers and beta blockers may increase the likelihood of seizures by inducing excessive excitability in the brain, which can trigger seizures.

Other research presented at the meeting found that angiotensin receptor blockers reduce the risk for epilepsy in patients with hypertension even in the absence of stroke.

Using data from an US national administrative database, the retrospective cohort study included 2,261,964 eligible adult beneficiaries diagnosed with hypertension and dispensed at least one angiotensin receptor blocker, ACE inhibitor, beta blocker, or calcium channel blocker from 2010 to 2017.

The study found angiotensin receptor blockers were associated with lower epilepsy incidence than ACE inhibitors (hazard ratio [HR], 0.75; 95% CI, 0.58-0.96), beta blockers (HR, 0.70; 95% CI, 0.54-0.90), and calcium channel blockers (HR, 0.80; 95% CI, 0.61-1.04).

The effect was most robust for the angiotensin receptor blocker losartan and among patients with no preexisting stroke or cardiovascular conditions.

Researchers ran the analysis excluding stroke and the association was still there, said lead study author Kimford Meador, MD, professor of neurology and neurological sciences, Stanford University, Palo Alto, California. “People with hypertension who use angiotensin receptor blockers — even if they don’t have a stroke — are less likely to develop epilepsy than if they use other major anti-hypertensives.”

 

Exciting and Suggestive

These new studies are “exciting” and “very suggestive,” said Meador. “We don’t have any true antiepileptic drugs; we have antiseizure medications. If we could reduce the incidence of epilepsy, that would be a very big thing.”

But the research to date hasn’t been “definitive” and is subject to bias, he said. “What we really need is a large randomized controlled trial to see if this is real because any observational study in humans has the potential for confounding from unmeasured variables.”

If such a study does show the effect is real, “that would change clinical practice,” said Meador.

As angiotensin receptor blockers have anti-inflammatory effects, researchers are now investigating whether this class of drugs might be useful in other diseases linked to inflammation such as multiple sclerosis, said Meador.

Commenting on the research, Alain Zingraff Looti, MD, assistant professor of neurology and public health sciences, Penn State College of Medicine, Hershey, Pennsylvania, said this new research is “important” although still at a “very preliminary stage.”

As the studies so far have all been observational, “they showed an association, but that doesn’t mean causality,” said Looti. These studies just looked at known potential confounders, “and there may be some unknown confounders that could explain the association,” he added.

But he acknowledged the results make sense from a biological standpoint. He noted that infusing angiotensin receptor blockers into rats causes a “cascade reaction” leading to reduced inflammation in the brain and then to less epilepsy. “Inflammation plays a major role in several models of epilepsy,” he added.

While these new results are important, Looti, too, would like to see a randomized controlled trial to confirm the findings.

Looti and Meador reported no relevant financial disclosures.

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

Angiotensin receptor blockers are more effective than other antihypertensive medications in reducing the risk for post-stroke epilepsy (PSE), new research showed.

Investigators found angiotensin receptor blockers are better at cutting the PSE risk in individuals with high blood pressure than angiotensin-converting enzyme (ACE) inhibitors, calcium channel blockers, and beta-blockers.

The results suggested angiotensin receptor blockers could be a frontline strategy to proactively forestall development of epilepsy in patients with a history of stroke and hypertension, said the study’s co-lead investigator Giacomo Evangelista, MD, PhD, a resident in neurology at the Epilepsy Center at “G. d’Annunzio” University of Chieti-Pescara, Italy.

The findings were presented at the American Epilepsy Society (AES) 78th Annual Meeting 2024.

 

Stroke and Seizures Tightly Linked

Stroke is the most common cause of seizures in patients older than 60 years. About 6%-8% of new epilepsy diagnoses in older adult patients are associated with a brain ischemic event.

Hypertension is among the most common risk factors for both epilepsy and stroke. The European Society of Cardiology recommends ACE inhibitors and angiotensin receptor blockers be considered first-line hypertension treatments.

Angiotensin receptor blockers seem to provide a protective effect in terms of seizures in the general population, but their possible preventive role in PSE has not been clear.

The retrospective, observational study included 528 patients (mean age, 71.4 years; 57.19% men) with hypertension and ischemic stroke without a history of epilepsy.

Researchers divided patients into those who developed PSE during the 2-year follow-up and those who didn’t. The main outcome was incidence of PSE associated with angiotensin receptor blocker therapy compared with other antihypertensive drug classes (ACE inhibitors, calcium channel blockers, and beta blockers).

Of the total, 7.2% of patients developed PSE. The study found patients treated with an angiotensin receptor blockers had a lower risk for PSE (P = .009). PSE incidence was higher among patients receiving calcium channel blockers (P = .019) and beta blockers (P = .008), but ACE inhibitors did not appear to affect PSE risk.

Further analysis showed that patients taking a beta blocker were 120% more likely to develop PSE, and those taking a calcium channel blocker were 110% more likely to develop PSE than those taking an angiotensin receptor blocker. The corresponding comparison was 65% for those taking an ACE inhibitor.

 

Potential Mechanisms

Angiotensin receptor blockers block the angiotensin II type 1 receptor, which may lead to numerous neuroprotective benefits such as improved blood flow to the brain and less neuroinflammation. The blockage may also reduce epilepsy severity and seizure frequency.

ACE inhibitors work in the same system as angiotensin receptor blockers but don’t bind directly to the angiotensin receptor. Experts believe these drugs may lead to inflammation that raises the risk for PSE.

Calcium channel blockers and beta blockers may increase the likelihood of seizures by inducing excessive excitability in the brain, which can trigger seizures.

Other research presented at the meeting found that angiotensin receptor blockers reduce the risk for epilepsy in patients with hypertension even in the absence of stroke.

Using data from an US national administrative database, the retrospective cohort study included 2,261,964 eligible adult beneficiaries diagnosed with hypertension and dispensed at least one angiotensin receptor blocker, ACE inhibitor, beta blocker, or calcium channel blocker from 2010 to 2017.

The study found angiotensin receptor blockers were associated with lower epilepsy incidence than ACE inhibitors (hazard ratio [HR], 0.75; 95% CI, 0.58-0.96), beta blockers (HR, 0.70; 95% CI, 0.54-0.90), and calcium channel blockers (HR, 0.80; 95% CI, 0.61-1.04).

The effect was most robust for the angiotensin receptor blocker losartan and among patients with no preexisting stroke or cardiovascular conditions.

Researchers ran the analysis excluding stroke and the association was still there, said lead study author Kimford Meador, MD, professor of neurology and neurological sciences, Stanford University, Palo Alto, California. “People with hypertension who use angiotensin receptor blockers — even if they don’t have a stroke — are less likely to develop epilepsy than if they use other major anti-hypertensives.”

 

Exciting and Suggestive

These new studies are “exciting” and “very suggestive,” said Meador. “We don’t have any true antiepileptic drugs; we have antiseizure medications. If we could reduce the incidence of epilepsy, that would be a very big thing.”

But the research to date hasn’t been “definitive” and is subject to bias, he said. “What we really need is a large randomized controlled trial to see if this is real because any observational study in humans has the potential for confounding from unmeasured variables.”

If such a study does show the effect is real, “that would change clinical practice,” said Meador.

As angiotensin receptor blockers have anti-inflammatory effects, researchers are now investigating whether this class of drugs might be useful in other diseases linked to inflammation such as multiple sclerosis, said Meador.

Commenting on the research, Alain Zingraff Looti, MD, assistant professor of neurology and public health sciences, Penn State College of Medicine, Hershey, Pennsylvania, said this new research is “important” although still at a “very preliminary stage.”

As the studies so far have all been observational, “they showed an association, but that doesn’t mean causality,” said Looti. These studies just looked at known potential confounders, “and there may be some unknown confounders that could explain the association,” he added.

But he acknowledged the results make sense from a biological standpoint. He noted that infusing angiotensin receptor blockers into rats causes a “cascade reaction” leading to reduced inflammation in the brain and then to less epilepsy. “Inflammation plays a major role in several models of epilepsy,” he added.

While these new results are important, Looti, too, would like to see a randomized controlled trial to confirm the findings.

Looti and Meador reported no relevant financial disclosures.

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

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM AES 2024

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Tue, 12/17/2024 - 16:13
Un-Gate On Date
Tue, 12/17/2024 - 16:13
Use ProPublica
CFC Schedule Remove Status
Tue, 12/17/2024 - 16:13
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
survey writer start date
Tue, 12/17/2024 - 16:13

‘Urgent Need’ to Examine Anti-Seizure Medication Pricing

Article Type
Changed
Tue, 12/17/2024 - 16:07

The median cost of brand-name anti-seizure medications (ASMs) in the United States almost doubled from 2013 to 2023, whereas generic ASM prices declined over the same period, results of a new study showed.

These findings highlight an “urgent need” to examine ASM pricing, said study investigator Pradeep Javarayee, MD, MBA, assistant professor, Department of Neurology, Medical College of Wisconsin, Milwaukee.

“The price disparity between generic and brand-name anti-seizure medications is growing,” said Javarayee. “Understanding these trends is essential for clinicians to make cost-conscious prescribing decisions and to advocate for policies that ensure equitable access to treatment for patients with epilepsy.” 

The findings were presented at American Epilepsy Society (AES) 78th Annual Meeting 2024.

 

Concerning Trends

Researchers analyzed prices of ASMs using National Average Drug Acquisition Cost (NADAC) data provided by the Centers for Medicare & Medicaid Services from November 2013 to July 2023.

A publicly available resource, NADAC is widely regarded as the most reliable benchmark for drug reimbursement in the United States, said Javarayee. It is based on the price/unit of Medicaid-covered outpatient drugs collected though monthly surveys of retail community pharmacies across the United States.

The database includes medication names, strengths, formulations, and National Drug Codes.

The study examined 23 US Food and Drug Administration–approved ASMs, encompassing 223 oral formulations, which included 112 brand-name and 111 generic products.

To account for inflation, researchers adjusted ASM prices using the Consumer Price Index for Medicinal Drugs–Seasonally Adjusted. They also explored the effects of the COVID-19 pandemic on drug pricing.

The study uncovered trends that Javarayee found concerning. For example, between 2013 and 2023, the average price of brand-name ASMs increased from $8.71 to $15.43, whereas the average price of generic ASMs decreased from $1.39 to $1.26.

Most generic ASMs remained in the low-cost range ($0-$0.25) during the study period, whereas the proportion of higher-priced brand-name ASMs ($10-$50) increased.

The number of brand-name ASMs with a mean price exceeding $15 rose from two (2013-2016) to six (2017-2019) and then to eight (2020-2023).

 

Unsustainable Price Increases

The study uncovered examples of what Javarayee called “extreme” price differences. For instance, matched brand-name and generic ASMs with price differences of 1000%-9999% increased from 32.88% in 2013-2016 to 41.43% in 2020-2023.

Among brand-name ASMs, cenobamate (Xcopri), which is approved in the United States for partial-onset seizures, had the highest mean price between 2020 and 2023 ($35.59). For generic ASMs, topiramate had the highest mean price in the same time period ($6.64).

Looking at formulation-based cost differences, the study found generic immediate-release formulations were significantly cheaper than extended-release or delayed-release counterparts, with cost differences reaching as high as 7751.20%.

The study also showed the COVID-19 pandemic led to a 24.4% increase in brand-name ASM prices and a 23.1% decrease in generic ASM prices.

The results underscore the need to raise awareness of drug pricing dynamics and call for targeted policy interventions to alleviate the rising cost burden on patients, said Javarayee.

He emphasized that such price increases are unsustainable. “Addressing these disparities is critical not only for improving patient adherence and outcomes but also for reducing the overall economic strain on the healthcare system.”

 

‘Tip of the Iceberg’

Reached for a comment, Tim Welty, a pharmacist in Des Moines, Iowa, who specializes in epilepsy drugs, agreed that the issue of medication costs needs to be addressed.

“I absolutely think physicians and others need to advocate for transparency in pricing,” he said. It’s not unusual for physician requests to have newer ASMs covered for their patients to be denied, he added.

Welty also noted Congress is currently considering bills that address the lack of transparency in the way medications are reimbursed.

This new study doesn’t reveal what patients are paying for their medications, he said. “These data tell me the national average of the cost to the pharmacy, but don’t tell me what the patient is actually paying, or what the insurance companies are actually paying for these medications.”

A “hot topic” in the industry is the role of pharmacy benefit managers, who are contracted by health insurance companies to manage medication portfolios, in influencing drug prices.

Drug pricing is a complicated topic. “There’s a lot more behind this; this is just like a tip of the iceberg,” said Welty.

Also commenting, Jacqueline French, MD, professor, NYU Comprehensive Epilepsy Center, New York City, said the much higher ASM prices are “very problematic,” particularly for drugs that don’t have a generic.

“I wonder how much the drugs for orphan indications, which are usually higher priced, influenced the numbers.”

The good news, though, is that many people “can safely take generics if they’re available,” said French.

The investigators and Welty reported no relevant disclosures.

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

Publications
Topics
Sections

The median cost of brand-name anti-seizure medications (ASMs) in the United States almost doubled from 2013 to 2023, whereas generic ASM prices declined over the same period, results of a new study showed.

These findings highlight an “urgent need” to examine ASM pricing, said study investigator Pradeep Javarayee, MD, MBA, assistant professor, Department of Neurology, Medical College of Wisconsin, Milwaukee.

“The price disparity between generic and brand-name anti-seizure medications is growing,” said Javarayee. “Understanding these trends is essential for clinicians to make cost-conscious prescribing decisions and to advocate for policies that ensure equitable access to treatment for patients with epilepsy.” 

The findings were presented at American Epilepsy Society (AES) 78th Annual Meeting 2024.

 

Concerning Trends

Researchers analyzed prices of ASMs using National Average Drug Acquisition Cost (NADAC) data provided by the Centers for Medicare & Medicaid Services from November 2013 to July 2023.

A publicly available resource, NADAC is widely regarded as the most reliable benchmark for drug reimbursement in the United States, said Javarayee. It is based on the price/unit of Medicaid-covered outpatient drugs collected though monthly surveys of retail community pharmacies across the United States.

The database includes medication names, strengths, formulations, and National Drug Codes.

The study examined 23 US Food and Drug Administration–approved ASMs, encompassing 223 oral formulations, which included 112 brand-name and 111 generic products.

To account for inflation, researchers adjusted ASM prices using the Consumer Price Index for Medicinal Drugs–Seasonally Adjusted. They also explored the effects of the COVID-19 pandemic on drug pricing.

The study uncovered trends that Javarayee found concerning. For example, between 2013 and 2023, the average price of brand-name ASMs increased from $8.71 to $15.43, whereas the average price of generic ASMs decreased from $1.39 to $1.26.

Most generic ASMs remained in the low-cost range ($0-$0.25) during the study period, whereas the proportion of higher-priced brand-name ASMs ($10-$50) increased.

The number of brand-name ASMs with a mean price exceeding $15 rose from two (2013-2016) to six (2017-2019) and then to eight (2020-2023).

 

Unsustainable Price Increases

The study uncovered examples of what Javarayee called “extreme” price differences. For instance, matched brand-name and generic ASMs with price differences of 1000%-9999% increased from 32.88% in 2013-2016 to 41.43% in 2020-2023.

Among brand-name ASMs, cenobamate (Xcopri), which is approved in the United States for partial-onset seizures, had the highest mean price between 2020 and 2023 ($35.59). For generic ASMs, topiramate had the highest mean price in the same time period ($6.64).

Looking at formulation-based cost differences, the study found generic immediate-release formulations were significantly cheaper than extended-release or delayed-release counterparts, with cost differences reaching as high as 7751.20%.

The study also showed the COVID-19 pandemic led to a 24.4% increase in brand-name ASM prices and a 23.1% decrease in generic ASM prices.

The results underscore the need to raise awareness of drug pricing dynamics and call for targeted policy interventions to alleviate the rising cost burden on patients, said Javarayee.

He emphasized that such price increases are unsustainable. “Addressing these disparities is critical not only for improving patient adherence and outcomes but also for reducing the overall economic strain on the healthcare system.”

 

‘Tip of the Iceberg’

Reached for a comment, Tim Welty, a pharmacist in Des Moines, Iowa, who specializes in epilepsy drugs, agreed that the issue of medication costs needs to be addressed.

“I absolutely think physicians and others need to advocate for transparency in pricing,” he said. It’s not unusual for physician requests to have newer ASMs covered for their patients to be denied, he added.

Welty also noted Congress is currently considering bills that address the lack of transparency in the way medications are reimbursed.

This new study doesn’t reveal what patients are paying for their medications, he said. “These data tell me the national average of the cost to the pharmacy, but don’t tell me what the patient is actually paying, or what the insurance companies are actually paying for these medications.”

A “hot topic” in the industry is the role of pharmacy benefit managers, who are contracted by health insurance companies to manage medication portfolios, in influencing drug prices.

Drug pricing is a complicated topic. “There’s a lot more behind this; this is just like a tip of the iceberg,” said Welty.

Also commenting, Jacqueline French, MD, professor, NYU Comprehensive Epilepsy Center, New York City, said the much higher ASM prices are “very problematic,” particularly for drugs that don’t have a generic.

“I wonder how much the drugs for orphan indications, which are usually higher priced, influenced the numbers.”

The good news, though, is that many people “can safely take generics if they’re available,” said French.

The investigators and Welty reported no relevant disclosures.

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

The median cost of brand-name anti-seizure medications (ASMs) in the United States almost doubled from 2013 to 2023, whereas generic ASM prices declined over the same period, results of a new study showed.

These findings highlight an “urgent need” to examine ASM pricing, said study investigator Pradeep Javarayee, MD, MBA, assistant professor, Department of Neurology, Medical College of Wisconsin, Milwaukee.

“The price disparity between generic and brand-name anti-seizure medications is growing,” said Javarayee. “Understanding these trends is essential for clinicians to make cost-conscious prescribing decisions and to advocate for policies that ensure equitable access to treatment for patients with epilepsy.” 

The findings were presented at American Epilepsy Society (AES) 78th Annual Meeting 2024.

 

Concerning Trends

Researchers analyzed prices of ASMs using National Average Drug Acquisition Cost (NADAC) data provided by the Centers for Medicare & Medicaid Services from November 2013 to July 2023.

A publicly available resource, NADAC is widely regarded as the most reliable benchmark for drug reimbursement in the United States, said Javarayee. It is based on the price/unit of Medicaid-covered outpatient drugs collected though monthly surveys of retail community pharmacies across the United States.

The database includes medication names, strengths, formulations, and National Drug Codes.

The study examined 23 US Food and Drug Administration–approved ASMs, encompassing 223 oral formulations, which included 112 brand-name and 111 generic products.

To account for inflation, researchers adjusted ASM prices using the Consumer Price Index for Medicinal Drugs–Seasonally Adjusted. They also explored the effects of the COVID-19 pandemic on drug pricing.

The study uncovered trends that Javarayee found concerning. For example, between 2013 and 2023, the average price of brand-name ASMs increased from $8.71 to $15.43, whereas the average price of generic ASMs decreased from $1.39 to $1.26.

Most generic ASMs remained in the low-cost range ($0-$0.25) during the study period, whereas the proportion of higher-priced brand-name ASMs ($10-$50) increased.

The number of brand-name ASMs with a mean price exceeding $15 rose from two (2013-2016) to six (2017-2019) and then to eight (2020-2023).

 

Unsustainable Price Increases

The study uncovered examples of what Javarayee called “extreme” price differences. For instance, matched brand-name and generic ASMs with price differences of 1000%-9999% increased from 32.88% in 2013-2016 to 41.43% in 2020-2023.

Among brand-name ASMs, cenobamate (Xcopri), which is approved in the United States for partial-onset seizures, had the highest mean price between 2020 and 2023 ($35.59). For generic ASMs, topiramate had the highest mean price in the same time period ($6.64).

Looking at formulation-based cost differences, the study found generic immediate-release formulations were significantly cheaper than extended-release or delayed-release counterparts, with cost differences reaching as high as 7751.20%.

The study also showed the COVID-19 pandemic led to a 24.4% increase in brand-name ASM prices and a 23.1% decrease in generic ASM prices.

The results underscore the need to raise awareness of drug pricing dynamics and call for targeted policy interventions to alleviate the rising cost burden on patients, said Javarayee.

He emphasized that such price increases are unsustainable. “Addressing these disparities is critical not only for improving patient adherence and outcomes but also for reducing the overall economic strain on the healthcare system.”

 

‘Tip of the Iceberg’

Reached for a comment, Tim Welty, a pharmacist in Des Moines, Iowa, who specializes in epilepsy drugs, agreed that the issue of medication costs needs to be addressed.

“I absolutely think physicians and others need to advocate for transparency in pricing,” he said. It’s not unusual for physician requests to have newer ASMs covered for their patients to be denied, he added.

Welty also noted Congress is currently considering bills that address the lack of transparency in the way medications are reimbursed.

This new study doesn’t reveal what patients are paying for their medications, he said. “These data tell me the national average of the cost to the pharmacy, but don’t tell me what the patient is actually paying, or what the insurance companies are actually paying for these medications.”

A “hot topic” in the industry is the role of pharmacy benefit managers, who are contracted by health insurance companies to manage medication portfolios, in influencing drug prices.

Drug pricing is a complicated topic. “There’s a lot more behind this; this is just like a tip of the iceberg,” said Welty.

Also commenting, Jacqueline French, MD, professor, NYU Comprehensive Epilepsy Center, New York City, said the much higher ASM prices are “very problematic,” particularly for drugs that don’t have a generic.

“I wonder how much the drugs for orphan indications, which are usually higher priced, influenced the numbers.”

The good news, though, is that many people “can safely take generics if they’re available,” said French.

The investigators and Welty reported no relevant disclosures.

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

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM AES 2024

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Tue, 12/17/2024 - 16:06
Un-Gate On Date
Tue, 12/17/2024 - 16:06
Use ProPublica
CFC Schedule Remove Status
Tue, 12/17/2024 - 16:06
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
survey writer start date
Tue, 12/17/2024 - 16:06

Is a Xanthine Oxidase Inhibitor or a Mechanism-Based Approach Best for First-Line Gout Treatment?

Article Type
Changed
Tue, 12/17/2024 - 15:18

For gout, xanthine oxidase (XO) inhibitors are the choice for first-line urate-lowering therapy (ULT) according to the 2020 American College of Rheumatology Guideline for the Management of Gout, which endorsed allopurinol, but should that be the approach for all patients, or should first-line therapy be tailored to the mechanism of each patient’s hyperuricemia? Two gout experts, Lisa Stamp, MBChB, PhD, a rheumatologist and professor of medicine at the University of Otago in Christchurch, New Zealand, and Fernando Pérez-Ruiz, MD, PhD, consultant in the Rheumatology Division of Cruces University Hospital, Barakaldo, head of the Investigation Group for Arthritis at Biocruces Health Research Institute, Barakaldo, and associate professor in the Department of Medicine at the Faculty of Medicine and Nursing at the University of the Basque Country in Leioa, Spain, debated this question recently at the annual research symposium of the Gout, Hyperuricemia and Crystal-Associated Disease Network.

Before the debate began, audience members voted on the question and 56% favored using XO inhibitors as a first-line therapy for all rather than tailoring first-line therapy to disease mechanism.

Up first, Stamp argued that XO inhibitors should be first-line therapy for all patients with gout. She said that XO inhibitors have been demonstrated to work regardless of the cause of hyperuricemia and degree of kidney function, and they are cheap, readily available, and easy to administer. She showed results from a study published by her debate opponent, Pérez-Ruiz, which demonstrated efficacy of XO inhibitors in both under-excreters and over-excreters of uric acid. That study compared the efficacy of the XO inhibitor allopurinol to the uricosuric agent benzbromarone and found the latter to be more effective, but Stamp argued that the allopurinol dose used in the study — 300 mg/d — “may not be enough for many patients who have gout. Dose-restricting allopurinol is one way to demonstrate that an alternative agent is superior,” she said.

A more recent study showed low-dose benzbromarone was better than low-dose febuxostat, another XO inhibitor. “I think we do need to have clinical trials that reflect real-world practice. I accept that this may have reflected [accepted practice where the studies] were undertaken, but these [XO inhibitor] doses don’t represent what many of us would do in other parts of the world,” Stamp said.

One concern is the utility of a ULT in patients with impaired renal function. Stamp cited her own post hoc analysis of a randomized, controlled trial showing that allopurinol is effective irrespective of renal function, as long as the dose is escalated to achieve target urate level, and a meta-analysis of observational studies suggesting that febuxostat is effective irrespective of renal function.

On the other hand, she showed data from a 1994 study of benzbromarone in renal transplant recipients, which showed that the drug’s effect on decreasing plasma uric acid dropped off significantly with lower creatinine clearance. “It does work, but the efficacy really drops off as renal function decreases. Benzbromarone is probably the most effective uricosuric in patients with renal impairment, but this agent is not readily available,” Stamp said.

The uricosuric agent probenecid, which is generally available across the world, led to only about 30% success in achieving target levels of uric acid among patients with an estimated glomerular filtration rate < 50 mL/min/1.73 m2. “I think we can all agree that getting 30% of our patients to target uric acid is not an acceptable outcome,” she said.

Stamp emphasized the importance of drug availability and noted that allopurinol is also the only medication for gout that is on the World Health Organization list of essential medications. “I think we should be recommending medications that are readily available, irrespective of where you live,” she said, noting that this is true of allopurinol, febuxostat, and probenecid.

Stamp also addressed the mechanism of action of ULTs. “Does the cause of hyperuricemia affect treatment response? I don’t think it does. Most people respond to allopurinol whether they’re a normal excreter or an under-excreter. Everyone who is lacking uricase will respond to allopurinol. Not everyone will respond to a uricosuric [agent], particularly in the setting of comorbidities such as renal impairment, which many of our patients with gout have,” she said.

 

Counterargument: Combine Therapies With Different Mechanisms of Action

In his counterargument, Pérez-Ruiz contended that gout is not in fact a metabolic disease and suggested that combining therapies with different mechanisms of action could be the best approach for difficult-to-treat gout. “The problem we face in clinical practice is how to treat difficult-to-treat patients,” he said. He referenced his own PhD thesis, which showed both high urinary uric acid output and underexcretion among patients with gout.

Pérez-Ruiz agreed that XO inhibitors should be used as first-line therapy but noted that the effect of allopurinol tapers off at higher doses. “If you use very high doses of allopurinol, you cannot expect to get much more effect,” he said. This is also true of febuxostat, he said.

He showed another study that illustrated difficulties in achieving target serum urate level with intensive therapy. “Even using a high dose of allopurinol, if you would like to get lower than 3 mg/dL for intensive therapy, close to 50% of patients will fail,” he said.

Pérez-Ruiz described a strategy of combining XO inhibitors with a uricosuric therapy, creating what he called a “uricase-like effect” on serum uric acid levels. Ruiz-Perez uses high-dose febuxostat in patients with chronic kidney disease who cannot be given uricosuric agents. “You can go to very low [serum urate levels] by raising up the doses,” he said.

He does not believe that allopurinol is the best agent for combination therapy in the treatment of tophaceous deposits. Instead, he favors combinations with febuxostat. He presented his own experience with 12 patients with very severe tophaceous gout who he treated with a combination of febuxostat and benzbromarone, which reduced serum urate to just over 2 mg/dL. “So this is a pegloticase-like effect [that is] very useful for tophaceous gout,” he said.

In her response, Stamp noted that most of the studies presented by Pérez-Ruiz showed XO inhibitors as first-line therapies, with other medications added on. “I think I heard Fernando agree with me. In just about all of those slides, he showed that a xanthine oxidase inhibitor was the first-line therapy, and subsequently a uricosuric was added,” she said.

Still, Stamp took issue with the idea that serum urate needs to get as low as Pérez-Ruiz advocated for. “What’s the risk associated with getting a serum urate to that level? I’m not sure that a sustained serum urate of around 1 [mg/dL] is necessarily good in the long term,” she said.

Stamp also pointed out the potential risks of polypharmacy, along with adherence issues. “If we can give our patients one therapy, one drug that’s going to get them to a target that we know is going to have beneficial long-term effects, that’s going to help improve our adherence. Maybe we are coming to a new era of [treatment, with] remission induction driving the serum urate very low, and then a maintenance therapy where we can back off. But irrespective, if you use that strategy, Fernando nicely showed that every time you’re going to start with a xanthine oxidase inhibitor,” she said.

After the debate, audience members voted again, and this time the result was 66% in favor of XO inhibitors as a first-line treatment.

Pérez-Ruiz is an adviser for Arthrosi, LG, Novartis, Protalix, and SOBI. He is a speaker for Menarini Central America and the Spanish Foundation for Rheumatology and has received funding from Cruces Rheumatology Association. Stamp did not disclose any financial relationships.

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

Meeting/Event
Publications
Topics
Sections
Meeting/Event
Meeting/Event

For gout, xanthine oxidase (XO) inhibitors are the choice for first-line urate-lowering therapy (ULT) according to the 2020 American College of Rheumatology Guideline for the Management of Gout, which endorsed allopurinol, but should that be the approach for all patients, or should first-line therapy be tailored to the mechanism of each patient’s hyperuricemia? Two gout experts, Lisa Stamp, MBChB, PhD, a rheumatologist and professor of medicine at the University of Otago in Christchurch, New Zealand, and Fernando Pérez-Ruiz, MD, PhD, consultant in the Rheumatology Division of Cruces University Hospital, Barakaldo, head of the Investigation Group for Arthritis at Biocruces Health Research Institute, Barakaldo, and associate professor in the Department of Medicine at the Faculty of Medicine and Nursing at the University of the Basque Country in Leioa, Spain, debated this question recently at the annual research symposium of the Gout, Hyperuricemia and Crystal-Associated Disease Network.

Before the debate began, audience members voted on the question and 56% favored using XO inhibitors as a first-line therapy for all rather than tailoring first-line therapy to disease mechanism.

Up first, Stamp argued that XO inhibitors should be first-line therapy for all patients with gout. She said that XO inhibitors have been demonstrated to work regardless of the cause of hyperuricemia and degree of kidney function, and they are cheap, readily available, and easy to administer. She showed results from a study published by her debate opponent, Pérez-Ruiz, which demonstrated efficacy of XO inhibitors in both under-excreters and over-excreters of uric acid. That study compared the efficacy of the XO inhibitor allopurinol to the uricosuric agent benzbromarone and found the latter to be more effective, but Stamp argued that the allopurinol dose used in the study — 300 mg/d — “may not be enough for many patients who have gout. Dose-restricting allopurinol is one way to demonstrate that an alternative agent is superior,” she said.

A more recent study showed low-dose benzbromarone was better than low-dose febuxostat, another XO inhibitor. “I think we do need to have clinical trials that reflect real-world practice. I accept that this may have reflected [accepted practice where the studies] were undertaken, but these [XO inhibitor] doses don’t represent what many of us would do in other parts of the world,” Stamp said.

One concern is the utility of a ULT in patients with impaired renal function. Stamp cited her own post hoc analysis of a randomized, controlled trial showing that allopurinol is effective irrespective of renal function, as long as the dose is escalated to achieve target urate level, and a meta-analysis of observational studies suggesting that febuxostat is effective irrespective of renal function.

On the other hand, she showed data from a 1994 study of benzbromarone in renal transplant recipients, which showed that the drug’s effect on decreasing plasma uric acid dropped off significantly with lower creatinine clearance. “It does work, but the efficacy really drops off as renal function decreases. Benzbromarone is probably the most effective uricosuric in patients with renal impairment, but this agent is not readily available,” Stamp said.

The uricosuric agent probenecid, which is generally available across the world, led to only about 30% success in achieving target levels of uric acid among patients with an estimated glomerular filtration rate < 50 mL/min/1.73 m2. “I think we can all agree that getting 30% of our patients to target uric acid is not an acceptable outcome,” she said.

Stamp emphasized the importance of drug availability and noted that allopurinol is also the only medication for gout that is on the World Health Organization list of essential medications. “I think we should be recommending medications that are readily available, irrespective of where you live,” she said, noting that this is true of allopurinol, febuxostat, and probenecid.

Stamp also addressed the mechanism of action of ULTs. “Does the cause of hyperuricemia affect treatment response? I don’t think it does. Most people respond to allopurinol whether they’re a normal excreter or an under-excreter. Everyone who is lacking uricase will respond to allopurinol. Not everyone will respond to a uricosuric [agent], particularly in the setting of comorbidities such as renal impairment, which many of our patients with gout have,” she said.

 

Counterargument: Combine Therapies With Different Mechanisms of Action

In his counterargument, Pérez-Ruiz contended that gout is not in fact a metabolic disease and suggested that combining therapies with different mechanisms of action could be the best approach for difficult-to-treat gout. “The problem we face in clinical practice is how to treat difficult-to-treat patients,” he said. He referenced his own PhD thesis, which showed both high urinary uric acid output and underexcretion among patients with gout.

Pérez-Ruiz agreed that XO inhibitors should be used as first-line therapy but noted that the effect of allopurinol tapers off at higher doses. “If you use very high doses of allopurinol, you cannot expect to get much more effect,” he said. This is also true of febuxostat, he said.

He showed another study that illustrated difficulties in achieving target serum urate level with intensive therapy. “Even using a high dose of allopurinol, if you would like to get lower than 3 mg/dL for intensive therapy, close to 50% of patients will fail,” he said.

Pérez-Ruiz described a strategy of combining XO inhibitors with a uricosuric therapy, creating what he called a “uricase-like effect” on serum uric acid levels. Ruiz-Perez uses high-dose febuxostat in patients with chronic kidney disease who cannot be given uricosuric agents. “You can go to very low [serum urate levels] by raising up the doses,” he said.

He does not believe that allopurinol is the best agent for combination therapy in the treatment of tophaceous deposits. Instead, he favors combinations with febuxostat. He presented his own experience with 12 patients with very severe tophaceous gout who he treated with a combination of febuxostat and benzbromarone, which reduced serum urate to just over 2 mg/dL. “So this is a pegloticase-like effect [that is] very useful for tophaceous gout,” he said.

In her response, Stamp noted that most of the studies presented by Pérez-Ruiz showed XO inhibitors as first-line therapies, with other medications added on. “I think I heard Fernando agree with me. In just about all of those slides, he showed that a xanthine oxidase inhibitor was the first-line therapy, and subsequently a uricosuric was added,” she said.

Still, Stamp took issue with the idea that serum urate needs to get as low as Pérez-Ruiz advocated for. “What’s the risk associated with getting a serum urate to that level? I’m not sure that a sustained serum urate of around 1 [mg/dL] is necessarily good in the long term,” she said.

Stamp also pointed out the potential risks of polypharmacy, along with adherence issues. “If we can give our patients one therapy, one drug that’s going to get them to a target that we know is going to have beneficial long-term effects, that’s going to help improve our adherence. Maybe we are coming to a new era of [treatment, with] remission induction driving the serum urate very low, and then a maintenance therapy where we can back off. But irrespective, if you use that strategy, Fernando nicely showed that every time you’re going to start with a xanthine oxidase inhibitor,” she said.

After the debate, audience members voted again, and this time the result was 66% in favor of XO inhibitors as a first-line treatment.

Pérez-Ruiz is an adviser for Arthrosi, LG, Novartis, Protalix, and SOBI. He is a speaker for Menarini Central America and the Spanish Foundation for Rheumatology and has received funding from Cruces Rheumatology Association. Stamp did not disclose any financial relationships.

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

For gout, xanthine oxidase (XO) inhibitors are the choice for first-line urate-lowering therapy (ULT) according to the 2020 American College of Rheumatology Guideline for the Management of Gout, which endorsed allopurinol, but should that be the approach for all patients, or should first-line therapy be tailored to the mechanism of each patient’s hyperuricemia? Two gout experts, Lisa Stamp, MBChB, PhD, a rheumatologist and professor of medicine at the University of Otago in Christchurch, New Zealand, and Fernando Pérez-Ruiz, MD, PhD, consultant in the Rheumatology Division of Cruces University Hospital, Barakaldo, head of the Investigation Group for Arthritis at Biocruces Health Research Institute, Barakaldo, and associate professor in the Department of Medicine at the Faculty of Medicine and Nursing at the University of the Basque Country in Leioa, Spain, debated this question recently at the annual research symposium of the Gout, Hyperuricemia and Crystal-Associated Disease Network.

Before the debate began, audience members voted on the question and 56% favored using XO inhibitors as a first-line therapy for all rather than tailoring first-line therapy to disease mechanism.

Up first, Stamp argued that XO inhibitors should be first-line therapy for all patients with gout. She said that XO inhibitors have been demonstrated to work regardless of the cause of hyperuricemia and degree of kidney function, and they are cheap, readily available, and easy to administer. She showed results from a study published by her debate opponent, Pérez-Ruiz, which demonstrated efficacy of XO inhibitors in both under-excreters and over-excreters of uric acid. That study compared the efficacy of the XO inhibitor allopurinol to the uricosuric agent benzbromarone and found the latter to be more effective, but Stamp argued that the allopurinol dose used in the study — 300 mg/d — “may not be enough for many patients who have gout. Dose-restricting allopurinol is one way to demonstrate that an alternative agent is superior,” she said.

A more recent study showed low-dose benzbromarone was better than low-dose febuxostat, another XO inhibitor. “I think we do need to have clinical trials that reflect real-world practice. I accept that this may have reflected [accepted practice where the studies] were undertaken, but these [XO inhibitor] doses don’t represent what many of us would do in other parts of the world,” Stamp said.

One concern is the utility of a ULT in patients with impaired renal function. Stamp cited her own post hoc analysis of a randomized, controlled trial showing that allopurinol is effective irrespective of renal function, as long as the dose is escalated to achieve target urate level, and a meta-analysis of observational studies suggesting that febuxostat is effective irrespective of renal function.

On the other hand, she showed data from a 1994 study of benzbromarone in renal transplant recipients, which showed that the drug’s effect on decreasing plasma uric acid dropped off significantly with lower creatinine clearance. “It does work, but the efficacy really drops off as renal function decreases. Benzbromarone is probably the most effective uricosuric in patients with renal impairment, but this agent is not readily available,” Stamp said.

The uricosuric agent probenecid, which is generally available across the world, led to only about 30% success in achieving target levels of uric acid among patients with an estimated glomerular filtration rate < 50 mL/min/1.73 m2. “I think we can all agree that getting 30% of our patients to target uric acid is not an acceptable outcome,” she said.

Stamp emphasized the importance of drug availability and noted that allopurinol is also the only medication for gout that is on the World Health Organization list of essential medications. “I think we should be recommending medications that are readily available, irrespective of where you live,” she said, noting that this is true of allopurinol, febuxostat, and probenecid.

Stamp also addressed the mechanism of action of ULTs. “Does the cause of hyperuricemia affect treatment response? I don’t think it does. Most people respond to allopurinol whether they’re a normal excreter or an under-excreter. Everyone who is lacking uricase will respond to allopurinol. Not everyone will respond to a uricosuric [agent], particularly in the setting of comorbidities such as renal impairment, which many of our patients with gout have,” she said.

 

Counterargument: Combine Therapies With Different Mechanisms of Action

In his counterargument, Pérez-Ruiz contended that gout is not in fact a metabolic disease and suggested that combining therapies with different mechanisms of action could be the best approach for difficult-to-treat gout. “The problem we face in clinical practice is how to treat difficult-to-treat patients,” he said. He referenced his own PhD thesis, which showed both high urinary uric acid output and underexcretion among patients with gout.

Pérez-Ruiz agreed that XO inhibitors should be used as first-line therapy but noted that the effect of allopurinol tapers off at higher doses. “If you use very high doses of allopurinol, you cannot expect to get much more effect,” he said. This is also true of febuxostat, he said.

He showed another study that illustrated difficulties in achieving target serum urate level with intensive therapy. “Even using a high dose of allopurinol, if you would like to get lower than 3 mg/dL for intensive therapy, close to 50% of patients will fail,” he said.

Pérez-Ruiz described a strategy of combining XO inhibitors with a uricosuric therapy, creating what he called a “uricase-like effect” on serum uric acid levels. Ruiz-Perez uses high-dose febuxostat in patients with chronic kidney disease who cannot be given uricosuric agents. “You can go to very low [serum urate levels] by raising up the doses,” he said.

He does not believe that allopurinol is the best agent for combination therapy in the treatment of tophaceous deposits. Instead, he favors combinations with febuxostat. He presented his own experience with 12 patients with very severe tophaceous gout who he treated with a combination of febuxostat and benzbromarone, which reduced serum urate to just over 2 mg/dL. “So this is a pegloticase-like effect [that is] very useful for tophaceous gout,” he said.

In her response, Stamp noted that most of the studies presented by Pérez-Ruiz showed XO inhibitors as first-line therapies, with other medications added on. “I think I heard Fernando agree with me. In just about all of those slides, he showed that a xanthine oxidase inhibitor was the first-line therapy, and subsequently a uricosuric was added,” she said.

Still, Stamp took issue with the idea that serum urate needs to get as low as Pérez-Ruiz advocated for. “What’s the risk associated with getting a serum urate to that level? I’m not sure that a sustained serum urate of around 1 [mg/dL] is necessarily good in the long term,” she said.

Stamp also pointed out the potential risks of polypharmacy, along with adherence issues. “If we can give our patients one therapy, one drug that’s going to get them to a target that we know is going to have beneficial long-term effects, that’s going to help improve our adherence. Maybe we are coming to a new era of [treatment, with] remission induction driving the serum urate very low, and then a maintenance therapy where we can back off. But irrespective, if you use that strategy, Fernando nicely showed that every time you’re going to start with a xanthine oxidase inhibitor,” she said.

After the debate, audience members voted again, and this time the result was 66% in favor of XO inhibitors as a first-line treatment.

Pérez-Ruiz is an adviser for Arthrosi, LG, Novartis, Protalix, and SOBI. He is a speaker for Menarini Central America and the Spanish Foundation for Rheumatology and has received funding from Cruces Rheumatology Association. Stamp did not disclose any financial relationships.

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

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM G-CAN 2024

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Tue, 12/17/2024 - 15:17
Un-Gate On Date
Tue, 12/17/2024 - 15:17
Use ProPublica
CFC Schedule Remove Status
Tue, 12/17/2024 - 15:17
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
survey writer start date
Tue, 12/17/2024 - 15:17

Daratumumab Confirmed as SOC for AL Amyloidosis

Article Type
Changed
Wed, 12/18/2024 - 06:42

— Final results of the phase 3 Andromeda study confirmed that adding daratumumab (DARA) — a human CD38-targeting monoclonal antibody — to bortezomib, cyclophosphamide, and dexamethasone (VCd) improves outcomes in patients with newly diagnosed amyloid light chain (AL) amyloidosis.

Adding DARA to VCd (D-VCd; Darzalex Faspro; Janssen Biotech) provided deeper and more rapid hematologic response and clinically meaningful and statistically significant improvement in overall survival (OS) and major organ deterioration progression-free survival (MOD-PFS), combined with 40.7% cardiac complete response (CR), first author Efstathios Kastritis, MD, said during presentation of an oral abstract at the American Society of Hematology (ASH) 2024 Annual Meeting.

“The Andromeda study is the first comparing two contemporary regimens that shows a significant survival improvement for patients with AL amyloidosis,” said Kastritis, an associate professor at the National and Kapodistrian University of Athens in Greece. “These findings reaffirm frontline D-VCd as the standard of care in this difficult-to-treat disease.”

The regimen was approved for this indication in 2021 based on prior earlier findings from the Andromeda trial. The current results are from a preplanned analysis for MOD-PFS and OS.

At a median follow-up of 61.4 months, the overall hematologic CR rates were 59.5% and 19.2% among 388 patients randomized to receive D-VCd or VCd, respectively (odds ratio, 6.03), which showed continued improvement with additional DARA vs the 53.3% and 18.1% rates observed at the primary analysis, Kastritis reported.

Time to hematologic CR was 67.5 days and 85.0 days in the treatment groups, respectively, and median duration of hematologic CR was not reached in either group.

A significant 56% improvement was also observed in MOD-PFS (hazard ratio [HR], 0.44). Median MOD-PFS was not reached in the D-VCd group and was 30.3 months in the VCd group.

A significant 38% improvement was observed in OS (HR, 0.62), 5-year OS was 76.1% vs 64.7% in the D-VCd and VCd groups, respectively, he said.

“[The OS] benefit occurred even though more than 70% of the patients in the VCd arm who received a subsequent therapy were treated with a DARA-based regimen,” he stressed. “This further emphasizes the importance of using DSTS in the frontline setting.”

Trial participants had newly diagnosed AL amyloidosis with measurable hematologic disease, one or more involved organs, cardiac stage I-IIIA, estimated glomerular filtration rate of at least 20 mL/min, and absence of symptomatic multiple myeloma. They were randomized 1:1 to the two treatment groups. All patients received 1.3 mg/m2 of bortezomib by weekly injection, 300 mg/m2 of cyclophosphamide either by weekly oral or intravenous administration, and 20-40 mg of dexamethasone by weekly oral or intravenous administration for six 28-day cycles.

Those in the D-VCd group also received 1800 mg of DARA coformulated with rHuPH20 as a weekly injection in cycles 1-2, as a biweekly injection in cycles 3-6, and by injection every 4 weeks thereafter for up to 24 28-day cycles.

The median duration of treatment was 21.3 months for D-VCd and 5.3 months for VCd, and of 122 patients who received subsequent therapy, 82 (67%) received subsequent DARA.

Patients who achieved hematologic CR had better MOD-PFS and OS (HR, 0.30 and 0.41, respectively), regardless of the treatment received, he noted, adding that “this further supports that complete hematologic response is a valid early endpoint for the evaluation of anti-monoclonal therapies in AL amyloidosis.”

“I think this is very important for the further development of new treatments in this disease,” he said.

Of note, cardiac and renal response rates in the D-VCd group were about two to three times greater than those in the VCd group at 6, 12, 24, 36, and 48 months, Kastritis said.

Among 235 patients with an evaluable cardiac response, 113 achieved a very good partial response or better, including 76 of 118 (64.4%) in the D-VCd group and 37 of 117 (31.6%) in the VCd group. Of these, 48 (40.7%) and 16 (13.7%) achieved a cardiac CR.

Grade 3 or 4 treatment-emergent adverse events (TEAEs) occurring in at least 5% of patients in the D-VCd and VCd groups, respectively, were lymphopenia (13% and 10%), pneumonia (8% and 4%), hypokalemia (2% and 5%), and peripheral edema (3% and 6%), he noted, adding that systemic administration-related reactions occurred in 14 (7%) of patients receiving D-VCd; all were grade 1 or 2 and most (86%) occurred after the first injection. TEAEs led to treatment discontinuation in 5% and 4% of patients in the groups, respectively.

No new safety signals were observed during the extended follow-up, he said.

Kastritis reported relationships with Pfizer, Genesis Pharma, Sanofi, AbbVie, GSK, Prothena, Janssen, and Amgen.

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

Meeting/Event
Publications
Topics
Sections
Meeting/Event
Meeting/Event

— Final results of the phase 3 Andromeda study confirmed that adding daratumumab (DARA) — a human CD38-targeting monoclonal antibody — to bortezomib, cyclophosphamide, and dexamethasone (VCd) improves outcomes in patients with newly diagnosed amyloid light chain (AL) amyloidosis.

Adding DARA to VCd (D-VCd; Darzalex Faspro; Janssen Biotech) provided deeper and more rapid hematologic response and clinically meaningful and statistically significant improvement in overall survival (OS) and major organ deterioration progression-free survival (MOD-PFS), combined with 40.7% cardiac complete response (CR), first author Efstathios Kastritis, MD, said during presentation of an oral abstract at the American Society of Hematology (ASH) 2024 Annual Meeting.

“The Andromeda study is the first comparing two contemporary regimens that shows a significant survival improvement for patients with AL amyloidosis,” said Kastritis, an associate professor at the National and Kapodistrian University of Athens in Greece. “These findings reaffirm frontline D-VCd as the standard of care in this difficult-to-treat disease.”

The regimen was approved for this indication in 2021 based on prior earlier findings from the Andromeda trial. The current results are from a preplanned analysis for MOD-PFS and OS.

At a median follow-up of 61.4 months, the overall hematologic CR rates were 59.5% and 19.2% among 388 patients randomized to receive D-VCd or VCd, respectively (odds ratio, 6.03), which showed continued improvement with additional DARA vs the 53.3% and 18.1% rates observed at the primary analysis, Kastritis reported.

Time to hematologic CR was 67.5 days and 85.0 days in the treatment groups, respectively, and median duration of hematologic CR was not reached in either group.

A significant 56% improvement was also observed in MOD-PFS (hazard ratio [HR], 0.44). Median MOD-PFS was not reached in the D-VCd group and was 30.3 months in the VCd group.

A significant 38% improvement was observed in OS (HR, 0.62), 5-year OS was 76.1% vs 64.7% in the D-VCd and VCd groups, respectively, he said.

“[The OS] benefit occurred even though more than 70% of the patients in the VCd arm who received a subsequent therapy were treated with a DARA-based regimen,” he stressed. “This further emphasizes the importance of using DSTS in the frontline setting.”

Trial participants had newly diagnosed AL amyloidosis with measurable hematologic disease, one or more involved organs, cardiac stage I-IIIA, estimated glomerular filtration rate of at least 20 mL/min, and absence of symptomatic multiple myeloma. They were randomized 1:1 to the two treatment groups. All patients received 1.3 mg/m2 of bortezomib by weekly injection, 300 mg/m2 of cyclophosphamide either by weekly oral or intravenous administration, and 20-40 mg of dexamethasone by weekly oral or intravenous administration for six 28-day cycles.

Those in the D-VCd group also received 1800 mg of DARA coformulated with rHuPH20 as a weekly injection in cycles 1-2, as a biweekly injection in cycles 3-6, and by injection every 4 weeks thereafter for up to 24 28-day cycles.

The median duration of treatment was 21.3 months for D-VCd and 5.3 months for VCd, and of 122 patients who received subsequent therapy, 82 (67%) received subsequent DARA.

Patients who achieved hematologic CR had better MOD-PFS and OS (HR, 0.30 and 0.41, respectively), regardless of the treatment received, he noted, adding that “this further supports that complete hematologic response is a valid early endpoint for the evaluation of anti-monoclonal therapies in AL amyloidosis.”

“I think this is very important for the further development of new treatments in this disease,” he said.

Of note, cardiac and renal response rates in the D-VCd group were about two to three times greater than those in the VCd group at 6, 12, 24, 36, and 48 months, Kastritis said.

Among 235 patients with an evaluable cardiac response, 113 achieved a very good partial response or better, including 76 of 118 (64.4%) in the D-VCd group and 37 of 117 (31.6%) in the VCd group. Of these, 48 (40.7%) and 16 (13.7%) achieved a cardiac CR.

Grade 3 or 4 treatment-emergent adverse events (TEAEs) occurring in at least 5% of patients in the D-VCd and VCd groups, respectively, were lymphopenia (13% and 10%), pneumonia (8% and 4%), hypokalemia (2% and 5%), and peripheral edema (3% and 6%), he noted, adding that systemic administration-related reactions occurred in 14 (7%) of patients receiving D-VCd; all were grade 1 or 2 and most (86%) occurred after the first injection. TEAEs led to treatment discontinuation in 5% and 4% of patients in the groups, respectively.

No new safety signals were observed during the extended follow-up, he said.

Kastritis reported relationships with Pfizer, Genesis Pharma, Sanofi, AbbVie, GSK, Prothena, Janssen, and Amgen.

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

— Final results of the phase 3 Andromeda study confirmed that adding daratumumab (DARA) — a human CD38-targeting monoclonal antibody — to bortezomib, cyclophosphamide, and dexamethasone (VCd) improves outcomes in patients with newly diagnosed amyloid light chain (AL) amyloidosis.

Adding DARA to VCd (D-VCd; Darzalex Faspro; Janssen Biotech) provided deeper and more rapid hematologic response and clinically meaningful and statistically significant improvement in overall survival (OS) and major organ deterioration progression-free survival (MOD-PFS), combined with 40.7% cardiac complete response (CR), first author Efstathios Kastritis, MD, said during presentation of an oral abstract at the American Society of Hematology (ASH) 2024 Annual Meeting.

“The Andromeda study is the first comparing two contemporary regimens that shows a significant survival improvement for patients with AL amyloidosis,” said Kastritis, an associate professor at the National and Kapodistrian University of Athens in Greece. “These findings reaffirm frontline D-VCd as the standard of care in this difficult-to-treat disease.”

The regimen was approved for this indication in 2021 based on prior earlier findings from the Andromeda trial. The current results are from a preplanned analysis for MOD-PFS and OS.

At a median follow-up of 61.4 months, the overall hematologic CR rates were 59.5% and 19.2% among 388 patients randomized to receive D-VCd or VCd, respectively (odds ratio, 6.03), which showed continued improvement with additional DARA vs the 53.3% and 18.1% rates observed at the primary analysis, Kastritis reported.

Time to hematologic CR was 67.5 days and 85.0 days in the treatment groups, respectively, and median duration of hematologic CR was not reached in either group.

A significant 56% improvement was also observed in MOD-PFS (hazard ratio [HR], 0.44). Median MOD-PFS was not reached in the D-VCd group and was 30.3 months in the VCd group.

A significant 38% improvement was observed in OS (HR, 0.62), 5-year OS was 76.1% vs 64.7% in the D-VCd and VCd groups, respectively, he said.

“[The OS] benefit occurred even though more than 70% of the patients in the VCd arm who received a subsequent therapy were treated with a DARA-based regimen,” he stressed. “This further emphasizes the importance of using DSTS in the frontline setting.”

Trial participants had newly diagnosed AL amyloidosis with measurable hematologic disease, one or more involved organs, cardiac stage I-IIIA, estimated glomerular filtration rate of at least 20 mL/min, and absence of symptomatic multiple myeloma. They were randomized 1:1 to the two treatment groups. All patients received 1.3 mg/m2 of bortezomib by weekly injection, 300 mg/m2 of cyclophosphamide either by weekly oral or intravenous administration, and 20-40 mg of dexamethasone by weekly oral or intravenous administration for six 28-day cycles.

Those in the D-VCd group also received 1800 mg of DARA coformulated with rHuPH20 as a weekly injection in cycles 1-2, as a biweekly injection in cycles 3-6, and by injection every 4 weeks thereafter for up to 24 28-day cycles.

The median duration of treatment was 21.3 months for D-VCd and 5.3 months for VCd, and of 122 patients who received subsequent therapy, 82 (67%) received subsequent DARA.

Patients who achieved hematologic CR had better MOD-PFS and OS (HR, 0.30 and 0.41, respectively), regardless of the treatment received, he noted, adding that “this further supports that complete hematologic response is a valid early endpoint for the evaluation of anti-monoclonal therapies in AL amyloidosis.”

“I think this is very important for the further development of new treatments in this disease,” he said.

Of note, cardiac and renal response rates in the D-VCd group were about two to three times greater than those in the VCd group at 6, 12, 24, 36, and 48 months, Kastritis said.

Among 235 patients with an evaluable cardiac response, 113 achieved a very good partial response or better, including 76 of 118 (64.4%) in the D-VCd group and 37 of 117 (31.6%) in the VCd group. Of these, 48 (40.7%) and 16 (13.7%) achieved a cardiac CR.

Grade 3 or 4 treatment-emergent adverse events (TEAEs) occurring in at least 5% of patients in the D-VCd and VCd groups, respectively, were lymphopenia (13% and 10%), pneumonia (8% and 4%), hypokalemia (2% and 5%), and peripheral edema (3% and 6%), he noted, adding that systemic administration-related reactions occurred in 14 (7%) of patients receiving D-VCd; all were grade 1 or 2 and most (86%) occurred after the first injection. TEAEs led to treatment discontinuation in 5% and 4% of patients in the groups, respectively.

No new safety signals were observed during the extended follow-up, he said.

Kastritis reported relationships with Pfizer, Genesis Pharma, Sanofi, AbbVie, GSK, Prothena, Janssen, and Amgen.

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

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM ASH 2024

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Tue, 12/17/2024 - 15:10
Un-Gate On Date
Tue, 12/17/2024 - 15:10
Use ProPublica
CFC Schedule Remove Status
Tue, 12/17/2024 - 15:10
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
survey writer start date
Tue, 12/17/2024 - 15:10

Upfront Therapy for ITP in Children: New Drug a Game-Changer?

Article Type
Changed
Wed, 12/18/2024 - 06:40

Children with newly diagnosed immune thrombocytopenia (ITP) treated with eltrombopag as a frontline therapy show significantly improved platelet responses and other outcomes compared with the standard of care.

“This is the first time in 30 years that a new drug is being tested for newly diagnosed pediatric ITP,” said the study’s lead author, Kristin A. Shimano, MD, professor of pediatrics at the Benioff Children’s Hospital, University of California San Francisco, in a press statement for the study, presented at the American Society of Hematology (ASH) 2024 Annual Meeting earlier this month.

“We really think that this has the potential to transform the approach to the management of ITP in the newly diagnosed phase with the use of a therapy that can provide sustained hemostatic platelet counts to bridge the time that patients are at risk of bleeding events with the goal to wean off the medication for patients who have a natural resolution of their disease,” Shimano said in her talk.

While children with ITP, a rare autoimmune blood disorder, very often improve without the need for any treatment, some do require intervention, and the condition can become chronic. First-line therapies for those patients commonly include corticosteroids, intravenous immunoglobulin (IVIg), and anti-D globulin; however, side effects can be undesirable, and with their efficacy often temporary, patients can require monitoring and juggling of treatments.

Eltrombopag, an oral, daily thrombopoietin receptor agonist, was approved by the US Food and Drug Administration for children and adults with chronic ITP in 2015; however, research has been lacking on the benefits of the therapy for newly diagnosed pediatric patients.

To investigate the drug’s efficacy at that stage, Shimano and colleagues with the ITP Consortium of North America launched the prospective, open-label Pediatric ITP Newly diagnosed pts Epag vs Standard therapy (PINES) trial, enrolling 118 patients at 23 institutions between May 2019 and January 2024.

All enrollees had been diagnosed with ITP within 3 months and had been determined by their treating hematologist to require pharmacologic treatment.

Of the patients, about 40% were untreated, and 60% had been treated with at least one medication prior to the trial but did not have a lasting response.

The patients were stratified by age and prior treatment and randomized 2:1 to receive either eltrombopag (n = 78) or the investigator’s choice of one of three standard first-line therapies, including prednisone, IVIg, or anti-D globulin at specified doses (n = 40). Overall, 29 in the standard-of-care arm received prednisone and 11 received IVIg. The patients had a median age of 8 years.

For the study’s primary endpoint, patients in the eltrombopag group had a significantly greater sustained response at 12 weeks, defined as having at least three of four platelet counts > 50 × 109/L during weeks 6-12 without the need for rescue treatment, with a rate of 63% vs 35% in the standard-of-care group (P = .0054).

There were no significant differences between the two groups in terms of the proportion of patients with a high bleeding score at weeks 1-4 and week 12.

However, those in the eltrombopag arm had a significantly lower rate of receiving rescue therapy (18% vs 38% with the standard of care; P = .02).

Both groups showed clinically meaningful improvements from baseline in terms of health-related quality of life, as assessed by parent proxy-reported KIT overall scores.

Twenty adverse events that were grade 3 or higher, including six serious adverse events, occurred in each of the study’s arms, with the most common events including headache and epistaxis.

Treatment-related serious adverse events occurred among six patients in the eltrombopag group and one in the control group, but importantly, no thromboembolic events were reported.

One intracranial hemorrhage occurred in the eltrombopag arm.

With eltrombopag having a slower effect than some other treatments, Shimano cautioned that the therapy is not recommended for patients with severe bleeding.

“Patients with grade 4 or 5 bleeding at the time of screening were specifically excluded from the study, so for patients who have very severe bleeding who need to get their platelets up very quickly, this would not be the ideal therapy for them,” she noted.

On the basis of results, the trial was recommended to close early due to efficacy; however, the participants are being followed for a total of 12 months to determine the durability of the responses, including in terms of bleeding events, quality of life, or the development of chronic ITP.

“We have shown that in pediatric patients with newly diagnosed ITP requiring pharmacologic treatment, eltrombopag resulted in a significant, clinically relevant higher rate of a durable platelet response in the absence of rescue treatment as compared with standard first-line therapies,” Shimano said.

“Eltrombopag could certainly be added to the medication choices hematologists consider as they are making treatment decisions with families, and it is an option that could potentially raise platelets for a more sustained period in children with ITP in the newly diagnosed period, which is one of the most difficult times for patients with regard to the impact of the disease on bleeding symptoms and quality of life,” she added.

Commenting on the study, James B. Bussel, MD, emeritus professor of pediatrics, medicine and obstetrics and gynecology at Weill Cornell Medicine in New York City, commented that “generally, a short-term increase in platelets is the biggest challenge, which is getting the patient to the point of not requiring future treatment to get better.”

“If more children can be shown to be going into remission earlier, that would be great,” he said.

While eltrombopag is known to be effective in chronic ITP, a key caveat of its use in newly diagnosed patients is the question of whether patients will get better on their own and feasibly be able to be spared the cost and burden of treatment in the first place.

However, identifying which patients will fit that profile isn’t always easy.

“Exactly which child needs treatment can be hard to determine, and there is some debate about that,” Bussel noted.

“The theoretic standard is that the platelet count doesn’t matter — only whether the patient is bleeding a lot, and then there is debate over treatment based on bleeding scores,” he said.

Quality-of-life issues, such as patients’ ability to take part in activities, are also a key consideration.

“It would be great if eltrombopag can support children who really need it and provide clear unequivocal benefit beyond just increasing the platelet count, but also leading to better quality of life,” Bussel said.

The new findings are “a very encouraging start, but I’d really like to see what the story is at 1 year.”

The study was funded by Novartis, maker of eltrombopag, and sponsored by the ITP Consortium of North America. Shimano disclosed ties with Sanofi, Sobi, Daiichi Sankyo, Novartis, and Pfizer. Bussel reported a relationship with Novartis that ended more than 2 years ago.

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

Meeting/Event
Publications
Topics
Sections
Meeting/Event
Meeting/Event

Children with newly diagnosed immune thrombocytopenia (ITP) treated with eltrombopag as a frontline therapy show significantly improved platelet responses and other outcomes compared with the standard of care.

“This is the first time in 30 years that a new drug is being tested for newly diagnosed pediatric ITP,” said the study’s lead author, Kristin A. Shimano, MD, professor of pediatrics at the Benioff Children’s Hospital, University of California San Francisco, in a press statement for the study, presented at the American Society of Hematology (ASH) 2024 Annual Meeting earlier this month.

“We really think that this has the potential to transform the approach to the management of ITP in the newly diagnosed phase with the use of a therapy that can provide sustained hemostatic platelet counts to bridge the time that patients are at risk of bleeding events with the goal to wean off the medication for patients who have a natural resolution of their disease,” Shimano said in her talk.

While children with ITP, a rare autoimmune blood disorder, very often improve without the need for any treatment, some do require intervention, and the condition can become chronic. First-line therapies for those patients commonly include corticosteroids, intravenous immunoglobulin (IVIg), and anti-D globulin; however, side effects can be undesirable, and with their efficacy often temporary, patients can require monitoring and juggling of treatments.

Eltrombopag, an oral, daily thrombopoietin receptor agonist, was approved by the US Food and Drug Administration for children and adults with chronic ITP in 2015; however, research has been lacking on the benefits of the therapy for newly diagnosed pediatric patients.

To investigate the drug’s efficacy at that stage, Shimano and colleagues with the ITP Consortium of North America launched the prospective, open-label Pediatric ITP Newly diagnosed pts Epag vs Standard therapy (PINES) trial, enrolling 118 patients at 23 institutions between May 2019 and January 2024.

All enrollees had been diagnosed with ITP within 3 months and had been determined by their treating hematologist to require pharmacologic treatment.

Of the patients, about 40% were untreated, and 60% had been treated with at least one medication prior to the trial but did not have a lasting response.

The patients were stratified by age and prior treatment and randomized 2:1 to receive either eltrombopag (n = 78) or the investigator’s choice of one of three standard first-line therapies, including prednisone, IVIg, or anti-D globulin at specified doses (n = 40). Overall, 29 in the standard-of-care arm received prednisone and 11 received IVIg. The patients had a median age of 8 years.

For the study’s primary endpoint, patients in the eltrombopag group had a significantly greater sustained response at 12 weeks, defined as having at least three of four platelet counts > 50 × 109/L during weeks 6-12 without the need for rescue treatment, with a rate of 63% vs 35% in the standard-of-care group (P = .0054).

There were no significant differences between the two groups in terms of the proportion of patients with a high bleeding score at weeks 1-4 and week 12.

However, those in the eltrombopag arm had a significantly lower rate of receiving rescue therapy (18% vs 38% with the standard of care; P = .02).

Both groups showed clinically meaningful improvements from baseline in terms of health-related quality of life, as assessed by parent proxy-reported KIT overall scores.

Twenty adverse events that were grade 3 or higher, including six serious adverse events, occurred in each of the study’s arms, with the most common events including headache and epistaxis.

Treatment-related serious adverse events occurred among six patients in the eltrombopag group and one in the control group, but importantly, no thromboembolic events were reported.

One intracranial hemorrhage occurred in the eltrombopag arm.

With eltrombopag having a slower effect than some other treatments, Shimano cautioned that the therapy is not recommended for patients with severe bleeding.

“Patients with grade 4 or 5 bleeding at the time of screening were specifically excluded from the study, so for patients who have very severe bleeding who need to get their platelets up very quickly, this would not be the ideal therapy for them,” she noted.

On the basis of results, the trial was recommended to close early due to efficacy; however, the participants are being followed for a total of 12 months to determine the durability of the responses, including in terms of bleeding events, quality of life, or the development of chronic ITP.

“We have shown that in pediatric patients with newly diagnosed ITP requiring pharmacologic treatment, eltrombopag resulted in a significant, clinically relevant higher rate of a durable platelet response in the absence of rescue treatment as compared with standard first-line therapies,” Shimano said.

“Eltrombopag could certainly be added to the medication choices hematologists consider as they are making treatment decisions with families, and it is an option that could potentially raise platelets for a more sustained period in children with ITP in the newly diagnosed period, which is one of the most difficult times for patients with regard to the impact of the disease on bleeding symptoms and quality of life,” she added.

Commenting on the study, James B. Bussel, MD, emeritus professor of pediatrics, medicine and obstetrics and gynecology at Weill Cornell Medicine in New York City, commented that “generally, a short-term increase in platelets is the biggest challenge, which is getting the patient to the point of not requiring future treatment to get better.”

“If more children can be shown to be going into remission earlier, that would be great,” he said.

While eltrombopag is known to be effective in chronic ITP, a key caveat of its use in newly diagnosed patients is the question of whether patients will get better on their own and feasibly be able to be spared the cost and burden of treatment in the first place.

However, identifying which patients will fit that profile isn’t always easy.

“Exactly which child needs treatment can be hard to determine, and there is some debate about that,” Bussel noted.

“The theoretic standard is that the platelet count doesn’t matter — only whether the patient is bleeding a lot, and then there is debate over treatment based on bleeding scores,” he said.

Quality-of-life issues, such as patients’ ability to take part in activities, are also a key consideration.

“It would be great if eltrombopag can support children who really need it and provide clear unequivocal benefit beyond just increasing the platelet count, but also leading to better quality of life,” Bussel said.

The new findings are “a very encouraging start, but I’d really like to see what the story is at 1 year.”

The study was funded by Novartis, maker of eltrombopag, and sponsored by the ITP Consortium of North America. Shimano disclosed ties with Sanofi, Sobi, Daiichi Sankyo, Novartis, and Pfizer. Bussel reported a relationship with Novartis that ended more than 2 years ago.

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

Children with newly diagnosed immune thrombocytopenia (ITP) treated with eltrombopag as a frontline therapy show significantly improved platelet responses and other outcomes compared with the standard of care.

“This is the first time in 30 years that a new drug is being tested for newly diagnosed pediatric ITP,” said the study’s lead author, Kristin A. Shimano, MD, professor of pediatrics at the Benioff Children’s Hospital, University of California San Francisco, in a press statement for the study, presented at the American Society of Hematology (ASH) 2024 Annual Meeting earlier this month.

“We really think that this has the potential to transform the approach to the management of ITP in the newly diagnosed phase with the use of a therapy that can provide sustained hemostatic platelet counts to bridge the time that patients are at risk of bleeding events with the goal to wean off the medication for patients who have a natural resolution of their disease,” Shimano said in her talk.

While children with ITP, a rare autoimmune blood disorder, very often improve without the need for any treatment, some do require intervention, and the condition can become chronic. First-line therapies for those patients commonly include corticosteroids, intravenous immunoglobulin (IVIg), and anti-D globulin; however, side effects can be undesirable, and with their efficacy often temporary, patients can require monitoring and juggling of treatments.

Eltrombopag, an oral, daily thrombopoietin receptor agonist, was approved by the US Food and Drug Administration for children and adults with chronic ITP in 2015; however, research has been lacking on the benefits of the therapy for newly diagnosed pediatric patients.

To investigate the drug’s efficacy at that stage, Shimano and colleagues with the ITP Consortium of North America launched the prospective, open-label Pediatric ITP Newly diagnosed pts Epag vs Standard therapy (PINES) trial, enrolling 118 patients at 23 institutions between May 2019 and January 2024.

All enrollees had been diagnosed with ITP within 3 months and had been determined by their treating hematologist to require pharmacologic treatment.

Of the patients, about 40% were untreated, and 60% had been treated with at least one medication prior to the trial but did not have a lasting response.

The patients were stratified by age and prior treatment and randomized 2:1 to receive either eltrombopag (n = 78) or the investigator’s choice of one of three standard first-line therapies, including prednisone, IVIg, or anti-D globulin at specified doses (n = 40). Overall, 29 in the standard-of-care arm received prednisone and 11 received IVIg. The patients had a median age of 8 years.

For the study’s primary endpoint, patients in the eltrombopag group had a significantly greater sustained response at 12 weeks, defined as having at least three of four platelet counts > 50 × 109/L during weeks 6-12 without the need for rescue treatment, with a rate of 63% vs 35% in the standard-of-care group (P = .0054).

There were no significant differences between the two groups in terms of the proportion of patients with a high bleeding score at weeks 1-4 and week 12.

However, those in the eltrombopag arm had a significantly lower rate of receiving rescue therapy (18% vs 38% with the standard of care; P = .02).

Both groups showed clinically meaningful improvements from baseline in terms of health-related quality of life, as assessed by parent proxy-reported KIT overall scores.

Twenty adverse events that were grade 3 or higher, including six serious adverse events, occurred in each of the study’s arms, with the most common events including headache and epistaxis.

Treatment-related serious adverse events occurred among six patients in the eltrombopag group and one in the control group, but importantly, no thromboembolic events were reported.

One intracranial hemorrhage occurred in the eltrombopag arm.

With eltrombopag having a slower effect than some other treatments, Shimano cautioned that the therapy is not recommended for patients with severe bleeding.

“Patients with grade 4 or 5 bleeding at the time of screening were specifically excluded from the study, so for patients who have very severe bleeding who need to get their platelets up very quickly, this would not be the ideal therapy for them,” she noted.

On the basis of results, the trial was recommended to close early due to efficacy; however, the participants are being followed for a total of 12 months to determine the durability of the responses, including in terms of bleeding events, quality of life, or the development of chronic ITP.

“We have shown that in pediatric patients with newly diagnosed ITP requiring pharmacologic treatment, eltrombopag resulted in a significant, clinically relevant higher rate of a durable platelet response in the absence of rescue treatment as compared with standard first-line therapies,” Shimano said.

“Eltrombopag could certainly be added to the medication choices hematologists consider as they are making treatment decisions with families, and it is an option that could potentially raise platelets for a more sustained period in children with ITP in the newly diagnosed period, which is one of the most difficult times for patients with regard to the impact of the disease on bleeding symptoms and quality of life,” she added.

Commenting on the study, James B. Bussel, MD, emeritus professor of pediatrics, medicine and obstetrics and gynecology at Weill Cornell Medicine in New York City, commented that “generally, a short-term increase in platelets is the biggest challenge, which is getting the patient to the point of not requiring future treatment to get better.”

“If more children can be shown to be going into remission earlier, that would be great,” he said.

While eltrombopag is known to be effective in chronic ITP, a key caveat of its use in newly diagnosed patients is the question of whether patients will get better on their own and feasibly be able to be spared the cost and burden of treatment in the first place.

However, identifying which patients will fit that profile isn’t always easy.

“Exactly which child needs treatment can be hard to determine, and there is some debate about that,” Bussel noted.

“The theoretic standard is that the platelet count doesn’t matter — only whether the patient is bleeding a lot, and then there is debate over treatment based on bleeding scores,” he said.

Quality-of-life issues, such as patients’ ability to take part in activities, are also a key consideration.

“It would be great if eltrombopag can support children who really need it and provide clear unequivocal benefit beyond just increasing the platelet count, but also leading to better quality of life,” Bussel said.

The new findings are “a very encouraging start, but I’d really like to see what the story is at 1 year.”

The study was funded by Novartis, maker of eltrombopag, and sponsored by the ITP Consortium of North America. Shimano disclosed ties with Sanofi, Sobi, Daiichi Sankyo, Novartis, and Pfizer. Bussel reported a relationship with Novartis that ended more than 2 years ago.

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

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM ASH 2024

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Tue, 12/17/2024 - 15:06
Un-Gate On Date
Tue, 12/17/2024 - 15:06
Use ProPublica
CFC Schedule Remove Status
Tue, 12/17/2024 - 15:06
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
survey writer start date
Tue, 12/17/2024 - 15:06

Surveillance Instead of Surgery for Low-Risk DCIS?

Article Type
Changed
Tue, 12/17/2024 - 13:50

— A large trial has begun to make the case for active surveillance as an alternative to immediate surgery for low-risk ductal carcinoma in situ (DCIS).

At 2 years, investigators on the COMET trial found no clinically meaningful difference in the rates of ipsilateral invasive breast cancer among women randomized to active surveillance vs standard upfront surgery with or without radiation.

The 2-year findings suggest that surveillance is safe in the short term.

“While these results are provocative, I don’t think they’re quite practice-changing yet,” said lead investigator Shelley Hwang, MD, a surgical breast oncologist at Duke University in Durham, North Carolina, who presented the findings at the San Antonio Breast Cancer Symposium (SABCS) 2024.

For one thing, it generally takes longer than 2 years for DCIS to convert to invasive cancer, so it will be important to wait for the planned analyses at 5, 7, and 10 years to make sure there isn’t an excess number of invasive breast cancers in the surveillance arm, Hwang said.

If the results prove durable, however, the findings will likely be “practice-changing” for women who were at least 40 years old and had grade 1 or 2 hormone receptor–positive DCIS at low risk for conversion, Hwang said.

The goal of active surveillance is to prevent unnecessary treatment. During surveillance, lesions are monitored for changes that indicate conversion to more advanced disease, at which point guideline-concordant care begins.

Although DCIS can convert to invasive breast cancer, this doesn’t always happen. As a result, upfront surgery and radiation aren’t necessary for some women.

The COMET trial aimed to determine the short-term safety of an active monitoring approach compared with guideline-concordant care in patients with low-risk DCIS.

The prospective, randomized noninferiority trial included women aged 40 years or older with a new diagnosis of HR–positive grade 1 or grade 2 DCIS without invasive cancer from 100 US Alliance Cancer Cooperative Group clinical trial sites.

In the trial, 484 women with DCIS were randomized to active surveillance — breast mammography and physical exam every 6 months — and 473 were randomized to standard upfront surgery with or without radiation. Overall, 15.7% of participants were Black and 75.0% were White.

Patients in either group could elect to have endocrine therapy, typically over a 5-year period (71% of women in the active monitoring group and 65.5% in the surgery group opted for endocrine therapy).

At 2 years, the cumulative rate of ipsilateral invasive breast cancer was 4.2% in the surveillance group vs 5.9% in the upfront surgery arm.

The study also included a planned per-protocol analysis among 673 patients who strictly followed the study protocol — 246 in the guideline-concordant care group who had received surgery by 6 months and 427 in the surveillance group who initiated the active monitoring protocol at 6 months.

With almost half of patients randomized to surgery declined to have it, which indicates that patients are interested in active monitoring, Hwang said.

At 2 years, the cumulative rate of invasive breast cancer was 3.1% in the active surveillance group vs 8.7% in the upfront surgery arm.

Among patients receiving endocrine therapy, the rate of invasive cancer was 7.15% in the surgery group and 3.21% in the surveillance arm.

Endocrine therapy “may have resulted in a reduced rate of invasive cancer in the active monitoring group,” the study authors noted.

These findings bring up the question of whether endocrine therapy might be just as good as surgery for low-risk DCIS, Hwang added. Given that one third of women undergo mastectomy for DCIS, “I think it’s not an inconsequential question,” Hwang said.

The findings, however, also suggest that surveillance sometimes leaves invasive cancer behind, Hwang explained. Nearly all invasive cancers in the surgery group were found during the initial operation , which may explain the slightly higher rates of invasive cancers in this group. Had the active monitoring group undergone surgery as well, the incidence of invasive cancer may have been the same in both arms, Hwang said.

However, when invasive cancers were removed, there were no significant differences in tumor size, node status, or tumor grade between the two groups, suggesting that there might not be a clinical penalty for delayed intervention with active monitoring, Hwang said.

With more than 10% of patients in the surgery group opting for mastectomy, compared with 1.8% in the active monitoring group, the active monitoring approach may not increase the likelihood of an eventual need for more extensive surgery, the COMET authors explained.

 

What Strategy Do Patients Prefer?

A companion analysis of patient-reported outcomes in COMET found no meaningful differences in quality of life, symptoms, or anxiety among patients who opted for surveillance over surgery. Results from questionnaires on quality of life, anxiety, depression, and breast cancer concerns were comparable between the two groups, with no evidence of a substantial impact of one approach over the other at 2 years.

“The results of this secondary analysis suggest that the lived experiences of individuals with low-risk DCIS are similar during early follow-up regardless of treatment allocation,” the COMET investigators concluded.

Overall, the findings from COMET provide reassuring short-term data, said Neil Iyengar, MD, a medical breast oncologist at Memorial Sloan Kettering Cancer Center in New York City.

DCIS is not an aggressive cancer, and it’s not going to invade any time soon, so patients have time to consider their options, Iyengar told Medscape Medical News.

The 2-year findings from COMET also help inform patient discussions. “I can tell patients if they decide not to have surgery what the likelihood is that they are going to convert into invasive cancer” after 2 years, he said.

COMET was published in JAMA, and the PRO analysis was published in JAMA Oncology to coincide with the study presentations.

COMET is funded by the Patient-Centered Outcomes Research Institute and others. Hwang is a consultant for Merck and on the advisory board of Clinetic, Exai Bio, and Havah Therapeutics. Iyengar is an advisor and/or researcher for AstraZeneca, Novartis, Pfizer, and other companies.

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

Publications
Topics
Sections

— A large trial has begun to make the case for active surveillance as an alternative to immediate surgery for low-risk ductal carcinoma in situ (DCIS).

At 2 years, investigators on the COMET trial found no clinically meaningful difference in the rates of ipsilateral invasive breast cancer among women randomized to active surveillance vs standard upfront surgery with or without radiation.

The 2-year findings suggest that surveillance is safe in the short term.

“While these results are provocative, I don’t think they’re quite practice-changing yet,” said lead investigator Shelley Hwang, MD, a surgical breast oncologist at Duke University in Durham, North Carolina, who presented the findings at the San Antonio Breast Cancer Symposium (SABCS) 2024.

For one thing, it generally takes longer than 2 years for DCIS to convert to invasive cancer, so it will be important to wait for the planned analyses at 5, 7, and 10 years to make sure there isn’t an excess number of invasive breast cancers in the surveillance arm, Hwang said.

If the results prove durable, however, the findings will likely be “practice-changing” for women who were at least 40 years old and had grade 1 or 2 hormone receptor–positive DCIS at low risk for conversion, Hwang said.

The goal of active surveillance is to prevent unnecessary treatment. During surveillance, lesions are monitored for changes that indicate conversion to more advanced disease, at which point guideline-concordant care begins.

Although DCIS can convert to invasive breast cancer, this doesn’t always happen. As a result, upfront surgery and radiation aren’t necessary for some women.

The COMET trial aimed to determine the short-term safety of an active monitoring approach compared with guideline-concordant care in patients with low-risk DCIS.

The prospective, randomized noninferiority trial included women aged 40 years or older with a new diagnosis of HR–positive grade 1 or grade 2 DCIS without invasive cancer from 100 US Alliance Cancer Cooperative Group clinical trial sites.

In the trial, 484 women with DCIS were randomized to active surveillance — breast mammography and physical exam every 6 months — and 473 were randomized to standard upfront surgery with or without radiation. Overall, 15.7% of participants were Black and 75.0% were White.

Patients in either group could elect to have endocrine therapy, typically over a 5-year period (71% of women in the active monitoring group and 65.5% in the surgery group opted for endocrine therapy).

At 2 years, the cumulative rate of ipsilateral invasive breast cancer was 4.2% in the surveillance group vs 5.9% in the upfront surgery arm.

The study also included a planned per-protocol analysis among 673 patients who strictly followed the study protocol — 246 in the guideline-concordant care group who had received surgery by 6 months and 427 in the surveillance group who initiated the active monitoring protocol at 6 months.

With almost half of patients randomized to surgery declined to have it, which indicates that patients are interested in active monitoring, Hwang said.

At 2 years, the cumulative rate of invasive breast cancer was 3.1% in the active surveillance group vs 8.7% in the upfront surgery arm.

Among patients receiving endocrine therapy, the rate of invasive cancer was 7.15% in the surgery group and 3.21% in the surveillance arm.

Endocrine therapy “may have resulted in a reduced rate of invasive cancer in the active monitoring group,” the study authors noted.

These findings bring up the question of whether endocrine therapy might be just as good as surgery for low-risk DCIS, Hwang added. Given that one third of women undergo mastectomy for DCIS, “I think it’s not an inconsequential question,” Hwang said.

The findings, however, also suggest that surveillance sometimes leaves invasive cancer behind, Hwang explained. Nearly all invasive cancers in the surgery group were found during the initial operation , which may explain the slightly higher rates of invasive cancers in this group. Had the active monitoring group undergone surgery as well, the incidence of invasive cancer may have been the same in both arms, Hwang said.

However, when invasive cancers were removed, there were no significant differences in tumor size, node status, or tumor grade between the two groups, suggesting that there might not be a clinical penalty for delayed intervention with active monitoring, Hwang said.

With more than 10% of patients in the surgery group opting for mastectomy, compared with 1.8% in the active monitoring group, the active monitoring approach may not increase the likelihood of an eventual need for more extensive surgery, the COMET authors explained.

 

What Strategy Do Patients Prefer?

A companion analysis of patient-reported outcomes in COMET found no meaningful differences in quality of life, symptoms, or anxiety among patients who opted for surveillance over surgery. Results from questionnaires on quality of life, anxiety, depression, and breast cancer concerns were comparable between the two groups, with no evidence of a substantial impact of one approach over the other at 2 years.

“The results of this secondary analysis suggest that the lived experiences of individuals with low-risk DCIS are similar during early follow-up regardless of treatment allocation,” the COMET investigators concluded.

Overall, the findings from COMET provide reassuring short-term data, said Neil Iyengar, MD, a medical breast oncologist at Memorial Sloan Kettering Cancer Center in New York City.

DCIS is not an aggressive cancer, and it’s not going to invade any time soon, so patients have time to consider their options, Iyengar told Medscape Medical News.

The 2-year findings from COMET also help inform patient discussions. “I can tell patients if they decide not to have surgery what the likelihood is that they are going to convert into invasive cancer” after 2 years, he said.

COMET was published in JAMA, and the PRO analysis was published in JAMA Oncology to coincide with the study presentations.

COMET is funded by the Patient-Centered Outcomes Research Institute and others. Hwang is a consultant for Merck and on the advisory board of Clinetic, Exai Bio, and Havah Therapeutics. Iyengar is an advisor and/or researcher for AstraZeneca, Novartis, Pfizer, and other companies.

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

— A large trial has begun to make the case for active surveillance as an alternative to immediate surgery for low-risk ductal carcinoma in situ (DCIS).

At 2 years, investigators on the COMET trial found no clinically meaningful difference in the rates of ipsilateral invasive breast cancer among women randomized to active surveillance vs standard upfront surgery with or without radiation.

The 2-year findings suggest that surveillance is safe in the short term.

“While these results are provocative, I don’t think they’re quite practice-changing yet,” said lead investigator Shelley Hwang, MD, a surgical breast oncologist at Duke University in Durham, North Carolina, who presented the findings at the San Antonio Breast Cancer Symposium (SABCS) 2024.

For one thing, it generally takes longer than 2 years for DCIS to convert to invasive cancer, so it will be important to wait for the planned analyses at 5, 7, and 10 years to make sure there isn’t an excess number of invasive breast cancers in the surveillance arm, Hwang said.

If the results prove durable, however, the findings will likely be “practice-changing” for women who were at least 40 years old and had grade 1 or 2 hormone receptor–positive DCIS at low risk for conversion, Hwang said.

The goal of active surveillance is to prevent unnecessary treatment. During surveillance, lesions are monitored for changes that indicate conversion to more advanced disease, at which point guideline-concordant care begins.

Although DCIS can convert to invasive breast cancer, this doesn’t always happen. As a result, upfront surgery and radiation aren’t necessary for some women.

The COMET trial aimed to determine the short-term safety of an active monitoring approach compared with guideline-concordant care in patients with low-risk DCIS.

The prospective, randomized noninferiority trial included women aged 40 years or older with a new diagnosis of HR–positive grade 1 or grade 2 DCIS without invasive cancer from 100 US Alliance Cancer Cooperative Group clinical trial sites.

In the trial, 484 women with DCIS were randomized to active surveillance — breast mammography and physical exam every 6 months — and 473 were randomized to standard upfront surgery with or without radiation. Overall, 15.7% of participants were Black and 75.0% were White.

Patients in either group could elect to have endocrine therapy, typically over a 5-year period (71% of women in the active monitoring group and 65.5% in the surgery group opted for endocrine therapy).

At 2 years, the cumulative rate of ipsilateral invasive breast cancer was 4.2% in the surveillance group vs 5.9% in the upfront surgery arm.

The study also included a planned per-protocol analysis among 673 patients who strictly followed the study protocol — 246 in the guideline-concordant care group who had received surgery by 6 months and 427 in the surveillance group who initiated the active monitoring protocol at 6 months.

With almost half of patients randomized to surgery declined to have it, which indicates that patients are interested in active monitoring, Hwang said.

At 2 years, the cumulative rate of invasive breast cancer was 3.1% in the active surveillance group vs 8.7% in the upfront surgery arm.

Among patients receiving endocrine therapy, the rate of invasive cancer was 7.15% in the surgery group and 3.21% in the surveillance arm.

Endocrine therapy “may have resulted in a reduced rate of invasive cancer in the active monitoring group,” the study authors noted.

These findings bring up the question of whether endocrine therapy might be just as good as surgery for low-risk DCIS, Hwang added. Given that one third of women undergo mastectomy for DCIS, “I think it’s not an inconsequential question,” Hwang said.

The findings, however, also suggest that surveillance sometimes leaves invasive cancer behind, Hwang explained. Nearly all invasive cancers in the surgery group were found during the initial operation , which may explain the slightly higher rates of invasive cancers in this group. Had the active monitoring group undergone surgery as well, the incidence of invasive cancer may have been the same in both arms, Hwang said.

However, when invasive cancers were removed, there were no significant differences in tumor size, node status, or tumor grade between the two groups, suggesting that there might not be a clinical penalty for delayed intervention with active monitoring, Hwang said.

With more than 10% of patients in the surgery group opting for mastectomy, compared with 1.8% in the active monitoring group, the active monitoring approach may not increase the likelihood of an eventual need for more extensive surgery, the COMET authors explained.

 

What Strategy Do Patients Prefer?

A companion analysis of patient-reported outcomes in COMET found no meaningful differences in quality of life, symptoms, or anxiety among patients who opted for surveillance over surgery. Results from questionnaires on quality of life, anxiety, depression, and breast cancer concerns were comparable between the two groups, with no evidence of a substantial impact of one approach over the other at 2 years.

“The results of this secondary analysis suggest that the lived experiences of individuals with low-risk DCIS are similar during early follow-up regardless of treatment allocation,” the COMET investigators concluded.

Overall, the findings from COMET provide reassuring short-term data, said Neil Iyengar, MD, a medical breast oncologist at Memorial Sloan Kettering Cancer Center in New York City.

DCIS is not an aggressive cancer, and it’s not going to invade any time soon, so patients have time to consider their options, Iyengar told Medscape Medical News.

The 2-year findings from COMET also help inform patient discussions. “I can tell patients if they decide not to have surgery what the likelihood is that they are going to convert into invasive cancer” after 2 years, he said.

COMET was published in JAMA, and the PRO analysis was published in JAMA Oncology to coincide with the study presentations.

COMET is funded by the Patient-Centered Outcomes Research Institute and others. Hwang is a consultant for Merck and on the advisory board of Clinetic, Exai Bio, and Havah Therapeutics. Iyengar is an advisor and/or researcher for AstraZeneca, Novartis, Pfizer, and other companies.

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

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM SABCS 2024

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Tue, 12/17/2024 - 13:49
Un-Gate On Date
Tue, 12/17/2024 - 13:49
Use ProPublica
CFC Schedule Remove Status
Tue, 12/17/2024 - 13:49
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
survey writer start date
Tue, 12/17/2024 - 13:49

Tamoxifen Reduces Risk for Invasive Recurrence of Ductal Carcinoma

Article Type
Changed
Mon, 12/16/2024 - 15:33

— For patients with so-called “good-risk” ductal carcinoma in situ (DCIS) who did not have radiation therapy after breast-conserving surgery, adjuvant tamoxifen reduced their overall risks for invasive recurrence, but not their risks for recurrence in either the same or contralateral breast.

These findings come from an exploratory analysis of combined data from two clinical trials. They suggest that, for this select group of patients, the choice to forgo radiation following definitive surgery may be an acceptable option, assuming that they follow a full course of endocrine therapy.

“In the absence of survival impact for adjuvant therapy, the decision to recommend radiation therapy or endocrine therapy should be part of a shared decision process, and I think that this data helps us provide clearer data points to our patients to help them make choices between endocrine therapy and radiation therapy in the setting of good-risk [ductal carcinoma in situ],” said Jean L. Wright, MD, from the University of North Carolina at Chapel Hill.

She presented the findings in an oral abstract session and media briefing at San Antonio Breast Cancer Symposium (SABCS) 2024.

 

Trial Results Combined

Wright and colleagues looked at pooled data from two clinical trials that enrolled patients with low- or intermediate-grade DCIS with tumor size no larger than 2.5 cm, grade 1 or 2 lesions, and with surgical margins ≥ 3 mm.

The trials included NRG/RTOG 9804, with 317 patients who fit the “good-risk category,” and ECOG-ACRIN E5194, which included a cohort of 561 patients that met the good-risk definition used for the exploratory analysis.

In each trial, tamoxifen use was optional, and choices were tracked. In the NRG/RTOG trial, 66% of patients used tamoxifen and 34% did not. The respective percentages in the ECOG/ACRIN trial were 30% and 70%.

The majority of patients were adherent to the 5-year prescribed course of tamoxifen, Wright said.

 

Analysis Details

In the combined data, the median age of patients who used tamoxifen vs who did not use tamoxifen was 58 vs 61 years.

In all, 23% of women in both the tamoxifen yes or no groups were premenopausal, with the remainder either postmenopausal or of unknown menopausal status.

After a median follow-up of 14.85 years, the rate of 15-year ipsilateral breast recurrence (IBR) was 19% for patients who did not receive tamoxifen vs 11.4% for those who did. This translated into a hazard ratio for IBR on tamoxifen of 0.52 (P = .001).

Tamoxifen also reduced the risk for invasive recurrence in the same breast, with a 15-year invasive IBR rate of 11.5% in the no tamoxifen group vs 6% in the yes tamoxifen group.

However, as noted before, tamoxifen use was not associated with significant reduction in the risk for noninvasive DCIS recurrence in the same breast, as evidenced by a 15-year DCIS IBR rate of 8.1% without tamoxifen and 5.5% with tamoxifen, a difference that did not reach statistical significance.

 

A Surprising Result

One finding from the data that seemed to defy clinical wisdom was that tamoxifen use did not appear to significantly reduce the risk for events in the other breast. The 15-year rate of contralateral breast events was 8.8% in the no-tamoxifen group vs 5.6% in the yes tamoxifen group, a difference that was not statically significant.

“It was surprising that there was so little effect on contralateral disease,” commented Elinor Sawyer, MBBS, PhD, the invited discussant.

“But I think it’s really important, this decrease in ipsilateral invasive recurrence [with tamoxifen] because there are studies such as the Sloane study from the United Kingdom that show that if you develop an invasive recurrence after DCIS, you have a worse survival than those who develop a pure DCIS recurrence,” she said.

At the media briefing held prior to Wright’s presentation, moderator Virginia Kaklamani, MD, leader of the breast cancer program at UT Health San Antonio in Texas, also said that she found it surprising that tamoxifen did not reduce risk for contralateral breast cancer “since every study that we’ve done has shown that.”

“I also found that result a little bit surprising,” Wright agreed.

“I think the main feature that we want to focus on is that this was a group of patients with a very clear inclusion criteria of this good-risk DCIS, and even though the definition of good-risk DCIS included patients with [tumors] up to 2.5 cm in DCIS, we saw that, in reality, the patients enrolled had very small DCIS. So I’m wondering if it’s perhaps that it’s related to the fact that patients that were enrolled in these studies had really low-risk features and perhaps just had a lower risk of contralateral breast events as compared to a broader population of patients with DCIS,” she said.

The analysis by Wright and colleagues was supported by grants from the National Cancer Institute. Wright reported receiving honoraria from ASTRO and PER. Sawyer disclosed receiving grants/research support from Pfizer, Seagen, and IQIVIA. Kaklamani disclosed serving as a speaker and/or consultant for AstraZeneca, Celldex Therapeutics, Daiichi Sankyo, Genentech, Gilead, Lilly, Menarini, and Novartis and receiving research support from Eisai.

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

Meeting/Event
Publications
Topics
Sections
Meeting/Event
Meeting/Event

— For patients with so-called “good-risk” ductal carcinoma in situ (DCIS) who did not have radiation therapy after breast-conserving surgery, adjuvant tamoxifen reduced their overall risks for invasive recurrence, but not their risks for recurrence in either the same or contralateral breast.

These findings come from an exploratory analysis of combined data from two clinical trials. They suggest that, for this select group of patients, the choice to forgo radiation following definitive surgery may be an acceptable option, assuming that they follow a full course of endocrine therapy.

“In the absence of survival impact for adjuvant therapy, the decision to recommend radiation therapy or endocrine therapy should be part of a shared decision process, and I think that this data helps us provide clearer data points to our patients to help them make choices between endocrine therapy and radiation therapy in the setting of good-risk [ductal carcinoma in situ],” said Jean L. Wright, MD, from the University of North Carolina at Chapel Hill.

She presented the findings in an oral abstract session and media briefing at San Antonio Breast Cancer Symposium (SABCS) 2024.

 

Trial Results Combined

Wright and colleagues looked at pooled data from two clinical trials that enrolled patients with low- or intermediate-grade DCIS with tumor size no larger than 2.5 cm, grade 1 or 2 lesions, and with surgical margins ≥ 3 mm.

The trials included NRG/RTOG 9804, with 317 patients who fit the “good-risk category,” and ECOG-ACRIN E5194, which included a cohort of 561 patients that met the good-risk definition used for the exploratory analysis.

In each trial, tamoxifen use was optional, and choices were tracked. In the NRG/RTOG trial, 66% of patients used tamoxifen and 34% did not. The respective percentages in the ECOG/ACRIN trial were 30% and 70%.

The majority of patients were adherent to the 5-year prescribed course of tamoxifen, Wright said.

 

Analysis Details

In the combined data, the median age of patients who used tamoxifen vs who did not use tamoxifen was 58 vs 61 years.

In all, 23% of women in both the tamoxifen yes or no groups were premenopausal, with the remainder either postmenopausal or of unknown menopausal status.

After a median follow-up of 14.85 years, the rate of 15-year ipsilateral breast recurrence (IBR) was 19% for patients who did not receive tamoxifen vs 11.4% for those who did. This translated into a hazard ratio for IBR on tamoxifen of 0.52 (P = .001).

Tamoxifen also reduced the risk for invasive recurrence in the same breast, with a 15-year invasive IBR rate of 11.5% in the no tamoxifen group vs 6% in the yes tamoxifen group.

However, as noted before, tamoxifen use was not associated with significant reduction in the risk for noninvasive DCIS recurrence in the same breast, as evidenced by a 15-year DCIS IBR rate of 8.1% without tamoxifen and 5.5% with tamoxifen, a difference that did not reach statistical significance.

 

A Surprising Result

One finding from the data that seemed to defy clinical wisdom was that tamoxifen use did not appear to significantly reduce the risk for events in the other breast. The 15-year rate of contralateral breast events was 8.8% in the no-tamoxifen group vs 5.6% in the yes tamoxifen group, a difference that was not statically significant.

“It was surprising that there was so little effect on contralateral disease,” commented Elinor Sawyer, MBBS, PhD, the invited discussant.

“But I think it’s really important, this decrease in ipsilateral invasive recurrence [with tamoxifen] because there are studies such as the Sloane study from the United Kingdom that show that if you develop an invasive recurrence after DCIS, you have a worse survival than those who develop a pure DCIS recurrence,” she said.

At the media briefing held prior to Wright’s presentation, moderator Virginia Kaklamani, MD, leader of the breast cancer program at UT Health San Antonio in Texas, also said that she found it surprising that tamoxifen did not reduce risk for contralateral breast cancer “since every study that we’ve done has shown that.”

“I also found that result a little bit surprising,” Wright agreed.

“I think the main feature that we want to focus on is that this was a group of patients with a very clear inclusion criteria of this good-risk DCIS, and even though the definition of good-risk DCIS included patients with [tumors] up to 2.5 cm in DCIS, we saw that, in reality, the patients enrolled had very small DCIS. So I’m wondering if it’s perhaps that it’s related to the fact that patients that were enrolled in these studies had really low-risk features and perhaps just had a lower risk of contralateral breast events as compared to a broader population of patients with DCIS,” she said.

The analysis by Wright and colleagues was supported by grants from the National Cancer Institute. Wright reported receiving honoraria from ASTRO and PER. Sawyer disclosed receiving grants/research support from Pfizer, Seagen, and IQIVIA. Kaklamani disclosed serving as a speaker and/or consultant for AstraZeneca, Celldex Therapeutics, Daiichi Sankyo, Genentech, Gilead, Lilly, Menarini, and Novartis and receiving research support from Eisai.

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

— For patients with so-called “good-risk” ductal carcinoma in situ (DCIS) who did not have radiation therapy after breast-conserving surgery, adjuvant tamoxifen reduced their overall risks for invasive recurrence, but not their risks for recurrence in either the same or contralateral breast.

These findings come from an exploratory analysis of combined data from two clinical trials. They suggest that, for this select group of patients, the choice to forgo radiation following definitive surgery may be an acceptable option, assuming that they follow a full course of endocrine therapy.

“In the absence of survival impact for adjuvant therapy, the decision to recommend radiation therapy or endocrine therapy should be part of a shared decision process, and I think that this data helps us provide clearer data points to our patients to help them make choices between endocrine therapy and radiation therapy in the setting of good-risk [ductal carcinoma in situ],” said Jean L. Wright, MD, from the University of North Carolina at Chapel Hill.

She presented the findings in an oral abstract session and media briefing at San Antonio Breast Cancer Symposium (SABCS) 2024.

 

Trial Results Combined

Wright and colleagues looked at pooled data from two clinical trials that enrolled patients with low- or intermediate-grade DCIS with tumor size no larger than 2.5 cm, grade 1 or 2 lesions, and with surgical margins ≥ 3 mm.

The trials included NRG/RTOG 9804, with 317 patients who fit the “good-risk category,” and ECOG-ACRIN E5194, which included a cohort of 561 patients that met the good-risk definition used for the exploratory analysis.

In each trial, tamoxifen use was optional, and choices were tracked. In the NRG/RTOG trial, 66% of patients used tamoxifen and 34% did not. The respective percentages in the ECOG/ACRIN trial were 30% and 70%.

The majority of patients were adherent to the 5-year prescribed course of tamoxifen, Wright said.

 

Analysis Details

In the combined data, the median age of patients who used tamoxifen vs who did not use tamoxifen was 58 vs 61 years.

In all, 23% of women in both the tamoxifen yes or no groups were premenopausal, with the remainder either postmenopausal or of unknown menopausal status.

After a median follow-up of 14.85 years, the rate of 15-year ipsilateral breast recurrence (IBR) was 19% for patients who did not receive tamoxifen vs 11.4% for those who did. This translated into a hazard ratio for IBR on tamoxifen of 0.52 (P = .001).

Tamoxifen also reduced the risk for invasive recurrence in the same breast, with a 15-year invasive IBR rate of 11.5% in the no tamoxifen group vs 6% in the yes tamoxifen group.

However, as noted before, tamoxifen use was not associated with significant reduction in the risk for noninvasive DCIS recurrence in the same breast, as evidenced by a 15-year DCIS IBR rate of 8.1% without tamoxifen and 5.5% with tamoxifen, a difference that did not reach statistical significance.

 

A Surprising Result

One finding from the data that seemed to defy clinical wisdom was that tamoxifen use did not appear to significantly reduce the risk for events in the other breast. The 15-year rate of contralateral breast events was 8.8% in the no-tamoxifen group vs 5.6% in the yes tamoxifen group, a difference that was not statically significant.

“It was surprising that there was so little effect on contralateral disease,” commented Elinor Sawyer, MBBS, PhD, the invited discussant.

“But I think it’s really important, this decrease in ipsilateral invasive recurrence [with tamoxifen] because there are studies such as the Sloane study from the United Kingdom that show that if you develop an invasive recurrence after DCIS, you have a worse survival than those who develop a pure DCIS recurrence,” she said.

At the media briefing held prior to Wright’s presentation, moderator Virginia Kaklamani, MD, leader of the breast cancer program at UT Health San Antonio in Texas, also said that she found it surprising that tamoxifen did not reduce risk for contralateral breast cancer “since every study that we’ve done has shown that.”

“I also found that result a little bit surprising,” Wright agreed.

“I think the main feature that we want to focus on is that this was a group of patients with a very clear inclusion criteria of this good-risk DCIS, and even though the definition of good-risk DCIS included patients with [tumors] up to 2.5 cm in DCIS, we saw that, in reality, the patients enrolled had very small DCIS. So I’m wondering if it’s perhaps that it’s related to the fact that patients that were enrolled in these studies had really low-risk features and perhaps just had a lower risk of contralateral breast events as compared to a broader population of patients with DCIS,” she said.

The analysis by Wright and colleagues was supported by grants from the National Cancer Institute. Wright reported receiving honoraria from ASTRO and PER. Sawyer disclosed receiving grants/research support from Pfizer, Seagen, and IQIVIA. Kaklamani disclosed serving as a speaker and/or consultant for AstraZeneca, Celldex Therapeutics, Daiichi Sankyo, Genentech, Gilead, Lilly, Menarini, and Novartis and receiving research support from Eisai.

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

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM SABCS 2024

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Mon, 12/16/2024 - 15:32
Un-Gate On Date
Mon, 12/16/2024 - 15:32
Use ProPublica
CFC Schedule Remove Status
Mon, 12/16/2024 - 15:32
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
survey writer start date
Mon, 12/16/2024 - 15:32

Imlunestrant Shows PFS Benefit in Advanced Breast Cancer

Article Type
Changed
Mon, 12/16/2024 - 15:16

— Second-line treatment with imlunestrant improved progression-free survival compared with standard endocrine monotherapy in patients with advanced estrogen receptor (ER)–positive, human epidermal growth factor receptor 2 (HER2)–negative breast cancer who had ESR1 mutations, according to recent findings from the EMBER-3 trial.

This progression-free survival benefit with imlunestrant did not extend to the overall population, but a combination of imlunestrant plus abemaciclib did lead to a significant improvement in progression-free survival compared with imlunestrant alone, regardless of patients’ ESR1 mutation status.

Lead author Komal Jhaveri, MD, a breast oncologist at Memorial Sloan Kettering Cancer Center in New York City, called the findings “encouraging.”

The phase 3 results raise the possibility of a second-line all-oral targeted therapy option for patients with ER–positive, HER2–negative advanced breast cancer, said Jhaveri, who presented the findings at the San Antonio Breast Cancer Symposium (SABCS) 2024, which were published simultaneously in The New England Journal of Medicine.

However, outside experts provided a note of caution that the trial design may limit how relevant the findings are to clinical practice.

First-line treatment for advanced ER–positive, HER2–negative breast cancer includes an aromatase inhibitor, such as exemestane, and a CDK4/6 inhibitor, such as abemaciclib. However, an ESR1 mutation may develop, which can undermine the effectiveness of the aromatase inhibitor. These patients may swap in a selective estrogen receptor degrader (SERD) — typically, the injectable fulvestrant — in place of the aromatase inhibitor.

Over the past several years, researchers have searched for a better agent than fulvestrant because this injectable drug has limited efficacy in patients with ESR1 mutations, and the monthly intramuscular shots are painful and inconvenient for patients.

The oral SERD imlunestrant is one such candidate.

The EMBER-3 trial initially randomized 661 patients after progression/recurrence evenly to either imlunestrant monotherapy (400 mg once daily) or a standard treatment arm that included either exemestane or fulvestrant, with 90% of these patients receiving fulvestrant. Investigators added a third combination arm shortly after the trial started, which included 213 patients who received imlunestrant plus abemaciclib.

About 60% of the overall population had received prior CDK4/6 inhibitors, primarily palbociclib and ribociclib. About 37% of the study population had ESR1 mutations.

Among patients with ESR1 mutations, imlunestrant monotherapy led to a significant improvement in median progression-free survival of 5.5 months vs 3.8 months in the standard care arm (P < .001). Among all patients, however, progression-free survival was no different between the two arms — 5.6 months in the imlunestrant group vs 5.5 months in the standard care group.

When comparing the two treatment arms in the overall population, the median progression-free survival was significantly better in patients who received imlunestrant plus abemaciclib — 9.4 months vs 5.5 months in the imlunestrant group (hazard ratio [HR] for progression or death, 0.57; P < .001). The progression-free survival benefit held across most patient subgroups, regardless of ESR1 mutation status, as well as among patients who had received a CDK4/6 inhibitor previously.

Data from other studies presented at SABCS indicate that another SERD, elacestrant, in combination with abemaciclib, may provide a similar progression-free survival benefit in this patient population. Elacestrant was approved by the US Food and Drug Administration (FDA) in January 2023 for second-line treatment of patients with advanced breast cancer and ESR1 mutations.

The EMBER trial also reported early overall survival findings. Although immature, overall survival trends favored imlunestrant over the standard treatment. The estimated overall survival at 18 months was 77% in the imlunestrant group and 58.6% in the standard therapy group among patients with ESR1 mutations (HR, 0.55), and 78.6% in the imlunestrant group vs 71.8% in the standard-therapy group for all patients (HR, 0.69).

Common all-grade adverse events with imlunestrant vs standard therapy included fatigue (22.6% vs 13.3%), diarrhea (21.4% vs 11.7%), and nausea (17% vs 13%). Grade 3 or higher anemia and neutropenia were low and similar in both arms.

All-grade diarrhea (86%) and nausea (49%) were more common with the combination of imlunestrant and abemaciclib.

The incidence of grade 3 or higher events was 17% with imlunestrant monotherapy, 21% for standard treatment, and 49% for imlunestrant plus abemaciclib.

EMBER-3 discussant Harold Burstein, MD, PhD, a breast oncologist at the Dana-Farber Cancer Institute, Boston, said that, overall, oral SERDs are starting to “break out from the ESR1 mutation box,” perhaps reflecting the idea that an agent more active than fulvestrant in combination with a non-cross–resistant CDK4/6 inhibitor like abemaciclib might lead to a better long-term outcome, regardless of ESR1 status.

A major limit of EMBER-3, however, is that it did not compare imlunestrant/abemaciclib with fulvestrant/abemaciclib, which would have been a true standard-of-care control, said Burstein.

Kathy Miller, MD, a breast oncologist at Indiana University, Indianapolis, agreed.

She was also concerned about the use of monotherapy in the standard care arm.

“Monotherapy hormone therapy is not what people would be treated with,” Miller said. Patients would typically get fulvestrant with either a targeted therapy or everolimus.

Without appropriate controls, “the data are impossible to interpret” in the context of current practice, Miller told Medscape Medical News.

Eli Lilly, maker of imlunestrant, funded, designed, and largely conducted the trial. Jhaveri is a consultant and researcher for the company. Burstein and Miller had no disclosures. Miller is an editorial advisor for Medscape Oncology.
 

A version of this article appeared on Medscape.com.

Publications
Topics
Sections

— Second-line treatment with imlunestrant improved progression-free survival compared with standard endocrine monotherapy in patients with advanced estrogen receptor (ER)–positive, human epidermal growth factor receptor 2 (HER2)–negative breast cancer who had ESR1 mutations, according to recent findings from the EMBER-3 trial.

This progression-free survival benefit with imlunestrant did not extend to the overall population, but a combination of imlunestrant plus abemaciclib did lead to a significant improvement in progression-free survival compared with imlunestrant alone, regardless of patients’ ESR1 mutation status.

Lead author Komal Jhaveri, MD, a breast oncologist at Memorial Sloan Kettering Cancer Center in New York City, called the findings “encouraging.”

The phase 3 results raise the possibility of a second-line all-oral targeted therapy option for patients with ER–positive, HER2–negative advanced breast cancer, said Jhaveri, who presented the findings at the San Antonio Breast Cancer Symposium (SABCS) 2024, which were published simultaneously in The New England Journal of Medicine.

However, outside experts provided a note of caution that the trial design may limit how relevant the findings are to clinical practice.

First-line treatment for advanced ER–positive, HER2–negative breast cancer includes an aromatase inhibitor, such as exemestane, and a CDK4/6 inhibitor, such as abemaciclib. However, an ESR1 mutation may develop, which can undermine the effectiveness of the aromatase inhibitor. These patients may swap in a selective estrogen receptor degrader (SERD) — typically, the injectable fulvestrant — in place of the aromatase inhibitor.

Over the past several years, researchers have searched for a better agent than fulvestrant because this injectable drug has limited efficacy in patients with ESR1 mutations, and the monthly intramuscular shots are painful and inconvenient for patients.

The oral SERD imlunestrant is one such candidate.

The EMBER-3 trial initially randomized 661 patients after progression/recurrence evenly to either imlunestrant monotherapy (400 mg once daily) or a standard treatment arm that included either exemestane or fulvestrant, with 90% of these patients receiving fulvestrant. Investigators added a third combination arm shortly after the trial started, which included 213 patients who received imlunestrant plus abemaciclib.

About 60% of the overall population had received prior CDK4/6 inhibitors, primarily palbociclib and ribociclib. About 37% of the study population had ESR1 mutations.

Among patients with ESR1 mutations, imlunestrant monotherapy led to a significant improvement in median progression-free survival of 5.5 months vs 3.8 months in the standard care arm (P < .001). Among all patients, however, progression-free survival was no different between the two arms — 5.6 months in the imlunestrant group vs 5.5 months in the standard care group.

When comparing the two treatment arms in the overall population, the median progression-free survival was significantly better in patients who received imlunestrant plus abemaciclib — 9.4 months vs 5.5 months in the imlunestrant group (hazard ratio [HR] for progression or death, 0.57; P < .001). The progression-free survival benefit held across most patient subgroups, regardless of ESR1 mutation status, as well as among patients who had received a CDK4/6 inhibitor previously.

Data from other studies presented at SABCS indicate that another SERD, elacestrant, in combination with abemaciclib, may provide a similar progression-free survival benefit in this patient population. Elacestrant was approved by the US Food and Drug Administration (FDA) in January 2023 for second-line treatment of patients with advanced breast cancer and ESR1 mutations.

The EMBER trial also reported early overall survival findings. Although immature, overall survival trends favored imlunestrant over the standard treatment. The estimated overall survival at 18 months was 77% in the imlunestrant group and 58.6% in the standard therapy group among patients with ESR1 mutations (HR, 0.55), and 78.6% in the imlunestrant group vs 71.8% in the standard-therapy group for all patients (HR, 0.69).

Common all-grade adverse events with imlunestrant vs standard therapy included fatigue (22.6% vs 13.3%), diarrhea (21.4% vs 11.7%), and nausea (17% vs 13%). Grade 3 or higher anemia and neutropenia were low and similar in both arms.

All-grade diarrhea (86%) and nausea (49%) were more common with the combination of imlunestrant and abemaciclib.

The incidence of grade 3 or higher events was 17% with imlunestrant monotherapy, 21% for standard treatment, and 49% for imlunestrant plus abemaciclib.

EMBER-3 discussant Harold Burstein, MD, PhD, a breast oncologist at the Dana-Farber Cancer Institute, Boston, said that, overall, oral SERDs are starting to “break out from the ESR1 mutation box,” perhaps reflecting the idea that an agent more active than fulvestrant in combination with a non-cross–resistant CDK4/6 inhibitor like abemaciclib might lead to a better long-term outcome, regardless of ESR1 status.

A major limit of EMBER-3, however, is that it did not compare imlunestrant/abemaciclib with fulvestrant/abemaciclib, which would have been a true standard-of-care control, said Burstein.

Kathy Miller, MD, a breast oncologist at Indiana University, Indianapolis, agreed.

She was also concerned about the use of monotherapy in the standard care arm.

“Monotherapy hormone therapy is not what people would be treated with,” Miller said. Patients would typically get fulvestrant with either a targeted therapy or everolimus.

Without appropriate controls, “the data are impossible to interpret” in the context of current practice, Miller told Medscape Medical News.

Eli Lilly, maker of imlunestrant, funded, designed, and largely conducted the trial. Jhaveri is a consultant and researcher for the company. Burstein and Miller had no disclosures. Miller is an editorial advisor for Medscape Oncology.
 

A version of this article appeared on Medscape.com.

— Second-line treatment with imlunestrant improved progression-free survival compared with standard endocrine monotherapy in patients with advanced estrogen receptor (ER)–positive, human epidermal growth factor receptor 2 (HER2)–negative breast cancer who had ESR1 mutations, according to recent findings from the EMBER-3 trial.

This progression-free survival benefit with imlunestrant did not extend to the overall population, but a combination of imlunestrant plus abemaciclib did lead to a significant improvement in progression-free survival compared with imlunestrant alone, regardless of patients’ ESR1 mutation status.

Lead author Komal Jhaveri, MD, a breast oncologist at Memorial Sloan Kettering Cancer Center in New York City, called the findings “encouraging.”

The phase 3 results raise the possibility of a second-line all-oral targeted therapy option for patients with ER–positive, HER2–negative advanced breast cancer, said Jhaveri, who presented the findings at the San Antonio Breast Cancer Symposium (SABCS) 2024, which were published simultaneously in The New England Journal of Medicine.

However, outside experts provided a note of caution that the trial design may limit how relevant the findings are to clinical practice.

First-line treatment for advanced ER–positive, HER2–negative breast cancer includes an aromatase inhibitor, such as exemestane, and a CDK4/6 inhibitor, such as abemaciclib. However, an ESR1 mutation may develop, which can undermine the effectiveness of the aromatase inhibitor. These patients may swap in a selective estrogen receptor degrader (SERD) — typically, the injectable fulvestrant — in place of the aromatase inhibitor.

Over the past several years, researchers have searched for a better agent than fulvestrant because this injectable drug has limited efficacy in patients with ESR1 mutations, and the monthly intramuscular shots are painful and inconvenient for patients.

The oral SERD imlunestrant is one such candidate.

The EMBER-3 trial initially randomized 661 patients after progression/recurrence evenly to either imlunestrant monotherapy (400 mg once daily) or a standard treatment arm that included either exemestane or fulvestrant, with 90% of these patients receiving fulvestrant. Investigators added a third combination arm shortly after the trial started, which included 213 patients who received imlunestrant plus abemaciclib.

About 60% of the overall population had received prior CDK4/6 inhibitors, primarily palbociclib and ribociclib. About 37% of the study population had ESR1 mutations.

Among patients with ESR1 mutations, imlunestrant monotherapy led to a significant improvement in median progression-free survival of 5.5 months vs 3.8 months in the standard care arm (P < .001). Among all patients, however, progression-free survival was no different between the two arms — 5.6 months in the imlunestrant group vs 5.5 months in the standard care group.

When comparing the two treatment arms in the overall population, the median progression-free survival was significantly better in patients who received imlunestrant plus abemaciclib — 9.4 months vs 5.5 months in the imlunestrant group (hazard ratio [HR] for progression or death, 0.57; P < .001). The progression-free survival benefit held across most patient subgroups, regardless of ESR1 mutation status, as well as among patients who had received a CDK4/6 inhibitor previously.

Data from other studies presented at SABCS indicate that another SERD, elacestrant, in combination with abemaciclib, may provide a similar progression-free survival benefit in this patient population. Elacestrant was approved by the US Food and Drug Administration (FDA) in January 2023 for second-line treatment of patients with advanced breast cancer and ESR1 mutations.

The EMBER trial also reported early overall survival findings. Although immature, overall survival trends favored imlunestrant over the standard treatment. The estimated overall survival at 18 months was 77% in the imlunestrant group and 58.6% in the standard therapy group among patients with ESR1 mutations (HR, 0.55), and 78.6% in the imlunestrant group vs 71.8% in the standard-therapy group for all patients (HR, 0.69).

Common all-grade adverse events with imlunestrant vs standard therapy included fatigue (22.6% vs 13.3%), diarrhea (21.4% vs 11.7%), and nausea (17% vs 13%). Grade 3 or higher anemia and neutropenia were low and similar in both arms.

All-grade diarrhea (86%) and nausea (49%) were more common with the combination of imlunestrant and abemaciclib.

The incidence of grade 3 or higher events was 17% with imlunestrant monotherapy, 21% for standard treatment, and 49% for imlunestrant plus abemaciclib.

EMBER-3 discussant Harold Burstein, MD, PhD, a breast oncologist at the Dana-Farber Cancer Institute, Boston, said that, overall, oral SERDs are starting to “break out from the ESR1 mutation box,” perhaps reflecting the idea that an agent more active than fulvestrant in combination with a non-cross–resistant CDK4/6 inhibitor like abemaciclib might lead to a better long-term outcome, regardless of ESR1 status.

A major limit of EMBER-3, however, is that it did not compare imlunestrant/abemaciclib with fulvestrant/abemaciclib, which would have been a true standard-of-care control, said Burstein.

Kathy Miller, MD, a breast oncologist at Indiana University, Indianapolis, agreed.

She was also concerned about the use of monotherapy in the standard care arm.

“Monotherapy hormone therapy is not what people would be treated with,” Miller said. Patients would typically get fulvestrant with either a targeted therapy or everolimus.

Without appropriate controls, “the data are impossible to interpret” in the context of current practice, Miller told Medscape Medical News.

Eli Lilly, maker of imlunestrant, funded, designed, and largely conducted the trial. Jhaveri is a consultant and researcher for the company. Burstein and Miller had no disclosures. Miller is an editorial advisor for Medscape Oncology.
 

A version of this article appeared on Medscape.com.

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM SABCS 2024

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Mon, 12/16/2024 - 15:14
Un-Gate On Date
Mon, 12/16/2024 - 15:14
Use ProPublica
CFC Schedule Remove Status
Mon, 12/16/2024 - 15:14
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
survey writer start date
Mon, 12/16/2024 - 15:14

Smoldering MM: Big Prevention Benefits With Daratumumab?

Article Type
Changed
Tue, 12/17/2024 - 06:17

— The multiple myeloma (MM) drug daratumumab (Darzalex), an anti-CD38 monoclonal antibody, dramatically reduced progression to active MM or death in patients with high-risk smoldering MM (SMM), a landmark randomized, open-label, phase 3 study found.

Among 390 patients with SMM (194 assigned to daratumumab and 196 to active monitoring), progression to active MM or death over a follow-up of 65.2 (0-76.6) months was 51% lower in the daratumumab group vs active monitoring (34.5% vs 50.5%, hazard ratio [HR], 0.49; 95% CI, 0.36-0.67; P < .0001), researchers reported at the American Society of Hematology (ASH) 2024 Annual Meeting and in a simultaneous publication in the New England Journal of Medicine.

Rahul Banerjee, MD, an assistant professor with the University of Washington and Fred Hutchinson Cancer Center, both in Seattle, who wasn’t involved with the research, said the study “is a big deal, and I suspect this will ultimately lead to an FDA [Food and Drug Administration] approval for daratumumab in this setting. If using daratumumab up-front can prevent further myeloma and therefore make patients live longer, this would be immediately adopted at many practices.”

As study first author Meletios Athanasios Dimopoulos, MD, of National and Kapodistrian University of Athens and Alexandra General Hospital in Greece, noted at a news briefing, SMM is common, affecting 0.5% of the population aged over 40, per a 2023 Iceland study.

“Standard practice is close follow-up without immediate intervention. However, this oftentimes ends in organ tissue damage, and hypercalcemia, bone lesions, renal impairment, and anemia,” Dimopoulos said.

According to him, researchers launched the AQUILA study in light of indications that daratumumab may benefit patients with intermediate- and high-risk SMM.

For the study, researchers recruited patients from 2017 to 2019 in 23 countries with confirmed high-risk SMM for ≤ 5 years (median age, 64 [31-86] years; 47%-49% men; 83% White).

In the daratumumab group, the drug was administered in 28-day cycles until cycle 39, 36 months, or disease progression, whichever came first (median treatment duration, 38 months [35 months]).

At 5 years, progression-free survival (PFS) — the primary endpoint — was 63.1% (daratumumab) and 40.8% (active monitoring). Researchers estimated 60-month PFS rates at 63.1% and 40.8%, respectively, and overall response rates were 63.4% vs 2.0% (P < .0001), respectively.

The 60-month overall survival rates were 93.0% and 86.9% (HR, 0.52; 95% CI, 0.27-0.98) with 15 deaths in the daratumumab and 26 in the active monitoring group.

“During the follow-up period, there was continuous improvement in favor of the daratumumab arm,” Dimopoulos said. “Even after treatment was discontinued at 3 years, or even at 5 or 6 years, there was a continuous benefit from treatment with daratumumab.”

By clinical cutoff in May 2024, 65% of patients taking daratumumab had finished 39 cycles/3 years of treatment vs 40.8% in the active monitoring group. Progressive disease was the most common reason that patients stopped treatment (21.8% and 41.8% of patients in the groups, respectively).

Grade 3/4 treatment-emergent adverse events (TEAEs) occurred in 40.4% (daratumumab) and 30.1% (active monitoring) of patients. The most common was hypertension (5.7% and 4.6%, respectively).

In the daratumumab group, 5.7% discontinued therapy because of TEAEs, which the researchers described as a “low” number, and fatal TEAEs were similar in both groups (1.0% and 2.0%, respectively).

Banerjee said that “one theoretical risk of using daratumumab monotherapy to treat perceived high-risk SMM is that if the patient actually has active multiple myeloma, you are undertreating them. For anyone with HR-SMM, active multiple myeloma must be completely ruled out. I always insist on both a PET-CT and a whole-body MRI to evaluate the bone marrow comprehensively.”

For now, Banerjee said, clinicians should wait for the US Food and Drug Administration approval before prescribing daratumumab for high-risk SMM.

Are there alternatives to reduce the risk for SMM turning into MM? “Generally, I advise close observation in most cases, but we do have clinical trials in this space,” Banerjee said. “Technically, it is possible to consider lenalidomide monotherapy in SMM based on the results of a large phase 3 study. But lenalidomide is expensive and has many side effects. Insurance companies often won’t cover it fully, and patients almost always have at least one side effect.”

Also, he added, “only half of patients saw their high-risk SMM disease burden drop. Lenalidomide also has a clear link to rare, delayed toxicities such as second primary malignancies, which makes us nervous.”

Janssen Pharmaceuticals, the maker of daratumumab, funded the study. Dimopoulos disclosed ties with Sanofi, Regeneron, Menarini, Takeda, GSK, BMS, Janssen Pharmaceuticals, BeiGene, Swixx, AstraZeneca, and Amgen. Banerjee disclosed ties with AbbVie, Adaptive, BMS, Caribou, Genentech/Roche, GSK, Karyopharm Therapeutics, Legend, Johnson & Johnson, Novartis, Pack, Pfizer, Prothena, Sanofi Pasteur, and SparkCures. Some other authors reported various and multiple disclosures, including ties with Janssen Pharmaceuticals.

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

Meeting/Event
Publications
Topics
Sections
Meeting/Event
Meeting/Event

— The multiple myeloma (MM) drug daratumumab (Darzalex), an anti-CD38 monoclonal antibody, dramatically reduced progression to active MM or death in patients with high-risk smoldering MM (SMM), a landmark randomized, open-label, phase 3 study found.

Among 390 patients with SMM (194 assigned to daratumumab and 196 to active monitoring), progression to active MM or death over a follow-up of 65.2 (0-76.6) months was 51% lower in the daratumumab group vs active monitoring (34.5% vs 50.5%, hazard ratio [HR], 0.49; 95% CI, 0.36-0.67; P < .0001), researchers reported at the American Society of Hematology (ASH) 2024 Annual Meeting and in a simultaneous publication in the New England Journal of Medicine.

Rahul Banerjee, MD, an assistant professor with the University of Washington and Fred Hutchinson Cancer Center, both in Seattle, who wasn’t involved with the research, said the study “is a big deal, and I suspect this will ultimately lead to an FDA [Food and Drug Administration] approval for daratumumab in this setting. If using daratumumab up-front can prevent further myeloma and therefore make patients live longer, this would be immediately adopted at many practices.”

As study first author Meletios Athanasios Dimopoulos, MD, of National and Kapodistrian University of Athens and Alexandra General Hospital in Greece, noted at a news briefing, SMM is common, affecting 0.5% of the population aged over 40, per a 2023 Iceland study.

“Standard practice is close follow-up without immediate intervention. However, this oftentimes ends in organ tissue damage, and hypercalcemia, bone lesions, renal impairment, and anemia,” Dimopoulos said.

According to him, researchers launched the AQUILA study in light of indications that daratumumab may benefit patients with intermediate- and high-risk SMM.

For the study, researchers recruited patients from 2017 to 2019 in 23 countries with confirmed high-risk SMM for ≤ 5 years (median age, 64 [31-86] years; 47%-49% men; 83% White).

In the daratumumab group, the drug was administered in 28-day cycles until cycle 39, 36 months, or disease progression, whichever came first (median treatment duration, 38 months [35 months]).

At 5 years, progression-free survival (PFS) — the primary endpoint — was 63.1% (daratumumab) and 40.8% (active monitoring). Researchers estimated 60-month PFS rates at 63.1% and 40.8%, respectively, and overall response rates were 63.4% vs 2.0% (P < .0001), respectively.

The 60-month overall survival rates were 93.0% and 86.9% (HR, 0.52; 95% CI, 0.27-0.98) with 15 deaths in the daratumumab and 26 in the active monitoring group.

“During the follow-up period, there was continuous improvement in favor of the daratumumab arm,” Dimopoulos said. “Even after treatment was discontinued at 3 years, or even at 5 or 6 years, there was a continuous benefit from treatment with daratumumab.”

By clinical cutoff in May 2024, 65% of patients taking daratumumab had finished 39 cycles/3 years of treatment vs 40.8% in the active monitoring group. Progressive disease was the most common reason that patients stopped treatment (21.8% and 41.8% of patients in the groups, respectively).

Grade 3/4 treatment-emergent adverse events (TEAEs) occurred in 40.4% (daratumumab) and 30.1% (active monitoring) of patients. The most common was hypertension (5.7% and 4.6%, respectively).

In the daratumumab group, 5.7% discontinued therapy because of TEAEs, which the researchers described as a “low” number, and fatal TEAEs were similar in both groups (1.0% and 2.0%, respectively).

Banerjee said that “one theoretical risk of using daratumumab monotherapy to treat perceived high-risk SMM is that if the patient actually has active multiple myeloma, you are undertreating them. For anyone with HR-SMM, active multiple myeloma must be completely ruled out. I always insist on both a PET-CT and a whole-body MRI to evaluate the bone marrow comprehensively.”

For now, Banerjee said, clinicians should wait for the US Food and Drug Administration approval before prescribing daratumumab for high-risk SMM.

Are there alternatives to reduce the risk for SMM turning into MM? “Generally, I advise close observation in most cases, but we do have clinical trials in this space,” Banerjee said. “Technically, it is possible to consider lenalidomide monotherapy in SMM based on the results of a large phase 3 study. But lenalidomide is expensive and has many side effects. Insurance companies often won’t cover it fully, and patients almost always have at least one side effect.”

Also, he added, “only half of patients saw their high-risk SMM disease burden drop. Lenalidomide also has a clear link to rare, delayed toxicities such as second primary malignancies, which makes us nervous.”

Janssen Pharmaceuticals, the maker of daratumumab, funded the study. Dimopoulos disclosed ties with Sanofi, Regeneron, Menarini, Takeda, GSK, BMS, Janssen Pharmaceuticals, BeiGene, Swixx, AstraZeneca, and Amgen. Banerjee disclosed ties with AbbVie, Adaptive, BMS, Caribou, Genentech/Roche, GSK, Karyopharm Therapeutics, Legend, Johnson & Johnson, Novartis, Pack, Pfizer, Prothena, Sanofi Pasteur, and SparkCures. Some other authors reported various and multiple disclosures, including ties with Janssen Pharmaceuticals.

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

— The multiple myeloma (MM) drug daratumumab (Darzalex), an anti-CD38 monoclonal antibody, dramatically reduced progression to active MM or death in patients with high-risk smoldering MM (SMM), a landmark randomized, open-label, phase 3 study found.

Among 390 patients with SMM (194 assigned to daratumumab and 196 to active monitoring), progression to active MM or death over a follow-up of 65.2 (0-76.6) months was 51% lower in the daratumumab group vs active monitoring (34.5% vs 50.5%, hazard ratio [HR], 0.49; 95% CI, 0.36-0.67; P < .0001), researchers reported at the American Society of Hematology (ASH) 2024 Annual Meeting and in a simultaneous publication in the New England Journal of Medicine.

Rahul Banerjee, MD, an assistant professor with the University of Washington and Fred Hutchinson Cancer Center, both in Seattle, who wasn’t involved with the research, said the study “is a big deal, and I suspect this will ultimately lead to an FDA [Food and Drug Administration] approval for daratumumab in this setting. If using daratumumab up-front can prevent further myeloma and therefore make patients live longer, this would be immediately adopted at many practices.”

As study first author Meletios Athanasios Dimopoulos, MD, of National and Kapodistrian University of Athens and Alexandra General Hospital in Greece, noted at a news briefing, SMM is common, affecting 0.5% of the population aged over 40, per a 2023 Iceland study.

“Standard practice is close follow-up without immediate intervention. However, this oftentimes ends in organ tissue damage, and hypercalcemia, bone lesions, renal impairment, and anemia,” Dimopoulos said.

According to him, researchers launched the AQUILA study in light of indications that daratumumab may benefit patients with intermediate- and high-risk SMM.

For the study, researchers recruited patients from 2017 to 2019 in 23 countries with confirmed high-risk SMM for ≤ 5 years (median age, 64 [31-86] years; 47%-49% men; 83% White).

In the daratumumab group, the drug was administered in 28-day cycles until cycle 39, 36 months, or disease progression, whichever came first (median treatment duration, 38 months [35 months]).

At 5 years, progression-free survival (PFS) — the primary endpoint — was 63.1% (daratumumab) and 40.8% (active monitoring). Researchers estimated 60-month PFS rates at 63.1% and 40.8%, respectively, and overall response rates were 63.4% vs 2.0% (P < .0001), respectively.

The 60-month overall survival rates were 93.0% and 86.9% (HR, 0.52; 95% CI, 0.27-0.98) with 15 deaths in the daratumumab and 26 in the active monitoring group.

“During the follow-up period, there was continuous improvement in favor of the daratumumab arm,” Dimopoulos said. “Even after treatment was discontinued at 3 years, or even at 5 or 6 years, there was a continuous benefit from treatment with daratumumab.”

By clinical cutoff in May 2024, 65% of patients taking daratumumab had finished 39 cycles/3 years of treatment vs 40.8% in the active monitoring group. Progressive disease was the most common reason that patients stopped treatment (21.8% and 41.8% of patients in the groups, respectively).

Grade 3/4 treatment-emergent adverse events (TEAEs) occurred in 40.4% (daratumumab) and 30.1% (active monitoring) of patients. The most common was hypertension (5.7% and 4.6%, respectively).

In the daratumumab group, 5.7% discontinued therapy because of TEAEs, which the researchers described as a “low” number, and fatal TEAEs were similar in both groups (1.0% and 2.0%, respectively).

Banerjee said that “one theoretical risk of using daratumumab monotherapy to treat perceived high-risk SMM is that if the patient actually has active multiple myeloma, you are undertreating them. For anyone with HR-SMM, active multiple myeloma must be completely ruled out. I always insist on both a PET-CT and a whole-body MRI to evaluate the bone marrow comprehensively.”

For now, Banerjee said, clinicians should wait for the US Food and Drug Administration approval before prescribing daratumumab for high-risk SMM.

Are there alternatives to reduce the risk for SMM turning into MM? “Generally, I advise close observation in most cases, but we do have clinical trials in this space,” Banerjee said. “Technically, it is possible to consider lenalidomide monotherapy in SMM based on the results of a large phase 3 study. But lenalidomide is expensive and has many side effects. Insurance companies often won’t cover it fully, and patients almost always have at least one side effect.”

Also, he added, “only half of patients saw their high-risk SMM disease burden drop. Lenalidomide also has a clear link to rare, delayed toxicities such as second primary malignancies, which makes us nervous.”

Janssen Pharmaceuticals, the maker of daratumumab, funded the study. Dimopoulos disclosed ties with Sanofi, Regeneron, Menarini, Takeda, GSK, BMS, Janssen Pharmaceuticals, BeiGene, Swixx, AstraZeneca, and Amgen. Banerjee disclosed ties with AbbVie, Adaptive, BMS, Caribou, Genentech/Roche, GSK, Karyopharm Therapeutics, Legend, Johnson & Johnson, Novartis, Pack, Pfizer, Prothena, Sanofi Pasteur, and SparkCures. Some other authors reported various and multiple disclosures, including ties with Janssen Pharmaceuticals.

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

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM ASH 2024

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Mon, 12/16/2024 - 11:52
Un-Gate On Date
Mon, 12/16/2024 - 11:52
Use ProPublica
CFC Schedule Remove Status
Mon, 12/16/2024 - 11:52
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
survey writer start date
Mon, 12/16/2024 - 11:52