AMA: Opioid prescriptions down since 2013

Article Type
Changed
Thu, 03/28/2019 - 14:36

 

Opioid prescriptions dropped by 22% over the last 5 years, with decreases seen in all 50 states, according to a new report from the American Medical Association’s opioid task force.

“The largest decrease in opioid prescriptions in 25 years reflects the fact that physicians and other health care professionals are increasingly judicious when prescribing opioids,” Patrice A. Harris, MD, chair of the task force, said in the report.

Providers dispensed almost 56 million fewer opioid prescriptions in 2017 than they did in 2013: The number of prescriptions fell from nearly 252 million in 2013 down to 196 million in 2017, the task force said.

Other signs of progress against the opioid epidemic include the increasing number of health care professionals registered for prescription drug monitoring programs: It has more than tripled, from 472,000 in 2014 to 1.55 million in 2017. Furthermore, the number of naloxone prescriptions in 2017 had more than doubled, from about 3,500 per week to 8,000, the task force reported.

“Unfortunately, deaths related to heroin and illicit fentanyl, and to prescription opioids, continue to rise. These statistics prove that decreasing prescriptions alone will not end the epidemic. We need well-designed initiatives that bring together public and private insurers, policymakers, public health infrastructure, and communities with the shared goal to improve access and coverage for comprehensive pain management and treatment for substance use disorders,” Dr. Harris said.

Publications
Topics
Sections

 

Opioid prescriptions dropped by 22% over the last 5 years, with decreases seen in all 50 states, according to a new report from the American Medical Association’s opioid task force.

“The largest decrease in opioid prescriptions in 25 years reflects the fact that physicians and other health care professionals are increasingly judicious when prescribing opioids,” Patrice A. Harris, MD, chair of the task force, said in the report.

Providers dispensed almost 56 million fewer opioid prescriptions in 2017 than they did in 2013: The number of prescriptions fell from nearly 252 million in 2013 down to 196 million in 2017, the task force said.

Other signs of progress against the opioid epidemic include the increasing number of health care professionals registered for prescription drug monitoring programs: It has more than tripled, from 472,000 in 2014 to 1.55 million in 2017. Furthermore, the number of naloxone prescriptions in 2017 had more than doubled, from about 3,500 per week to 8,000, the task force reported.

“Unfortunately, deaths related to heroin and illicit fentanyl, and to prescription opioids, continue to rise. These statistics prove that decreasing prescriptions alone will not end the epidemic. We need well-designed initiatives that bring together public and private insurers, policymakers, public health infrastructure, and communities with the shared goal to improve access and coverage for comprehensive pain management and treatment for substance use disorders,” Dr. Harris said.

 

Opioid prescriptions dropped by 22% over the last 5 years, with decreases seen in all 50 states, according to a new report from the American Medical Association’s opioid task force.

“The largest decrease in opioid prescriptions in 25 years reflects the fact that physicians and other health care professionals are increasingly judicious when prescribing opioids,” Patrice A. Harris, MD, chair of the task force, said in the report.

Providers dispensed almost 56 million fewer opioid prescriptions in 2017 than they did in 2013: The number of prescriptions fell from nearly 252 million in 2013 down to 196 million in 2017, the task force said.

Other signs of progress against the opioid epidemic include the increasing number of health care professionals registered for prescription drug monitoring programs: It has more than tripled, from 472,000 in 2014 to 1.55 million in 2017. Furthermore, the number of naloxone prescriptions in 2017 had more than doubled, from about 3,500 per week to 8,000, the task force reported.

“Unfortunately, deaths related to heroin and illicit fentanyl, and to prescription opioids, continue to rise. These statistics prove that decreasing prescriptions alone will not end the epidemic. We need well-designed initiatives that bring together public and private insurers, policymakers, public health infrastructure, and communities with the shared goal to improve access and coverage for comprehensive pain management and treatment for substance use disorders,” Dr. Harris said.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica

Checkpoint inhibitor shows promise in advanced squamous-cell carcinoma

Article Type
Changed
Mon, 01/14/2019 - 10:25

An immune checkpoint inhibitor that targets the PD-1 receptor has shown “robust” efficacy among patients with advanced cutaneous squamous-cell carcinoma, according to researchers.

A combined phase 1/phase 2 study, published in the New England Journal of Medicine and presented simultaneously at the annual meeting of the American Society of Clinical Oncology, looked at the effect of monoclonal antibody cemiplimab in an expansion cohort of 26 patients with locally-advanced or metastatic cutaneous squamous-cell carcinoma who were not eligible for surgery. The phase 2 component involved 59 patients with metastatic disease.

Patients were treated with intravenous cemiplimab every 2 weeks for 48 weeks in the phase 1 study, and up to 96 weeks – or until unacceptable toxicity or disease progression – in the phase 2 study.

In the phase 1 study, researchers saw a response rate of 50% and a 65% rate of durable disease control, after a median follow-up of 11 months (1.1-17). The median time to response was 2.3 months, and more than half the patients (54%) who showed a response maintained that response past 6 months.

In the phase 2 study in patients with metastatic disease, 47% responded to the treatment – 24 patients showed a partial response and 4 showed a complete response. Of those who responded, 61% showed durable disease control after a median follow-up of 7.9 months.

The median time to response in this group of patients was 1.9 months, and 57% of those who did respond still showed a response at 6 months. However neither median progression-free survival nor median overall survival had been reached at the point of data cut-off.

The treatment showed similar effects in patients with regional and distant metastatic disease.

Advanced cutaneous squamous-cell carcinoma was thought to be an ideal target for immunotherapy because the high mutation burden in the tumor meant it would be sensitive to effector T cell attack, wrote Michael R. Migden, MD, of the University of Texas MD Anderson Cancer Center, Houston, and his coauthors.

 

 


“In addition, the dramatically increased risk of cutaneous squamous-cell carcinoma among people with immunosuppression pointed to an important role for immune surveillance with this cancer,” the authors wrote.

In the phase 2 study, 29% of patients experienced a serious adverse event – including two cases of pneumonitis – and three patients (5%) discontinued treatment. There were three deaths due to adverse events: One patient died from pneumonia complications, one died in his sleep, and one patient died following hypercalcemia and deep vein thrombosis.

Aside from these, most adverse events were grade 1 or 2. Around one-quarter of patients experienced diarrhea (27%) or fatigue (24%), while the other most common adverse events were nausea (17%), constipation (15%) and rash (15%). The authors noted that these adverse events were similar to those seen in other PD-1 inhibitors.

“Our results are consistent with an emerging theme regarding the high efficacy of immune checkpoint blockade for the treatment of hypermutated cancers, since the mutation burden of cutaneous squamous-cell carcinoma is similar to that reported for advanced solid tumors with microsatellite instability,” the authors wrote.
 

 

Cemiplimab is now being tested in a phase 2 trial in patients with advanced basal cell carcinoma.

The study was supported by Regeneron Pharmaceuticals and Sanofi. Eight authors declared funding from Regeneron to conduct the trial. Ten authors were employees of Regeneron. Fifteen authors also declared funding and payments from pharmaceutical companies outside the submitted work. Four had nothing to disclose.

SOURCE: Migden M et al. NEJM, 2018; June 4. doi: 10.1056/NEJMoa1805131.

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

An immune checkpoint inhibitor that targets the PD-1 receptor has shown “robust” efficacy among patients with advanced cutaneous squamous-cell carcinoma, according to researchers.

A combined phase 1/phase 2 study, published in the New England Journal of Medicine and presented simultaneously at the annual meeting of the American Society of Clinical Oncology, looked at the effect of monoclonal antibody cemiplimab in an expansion cohort of 26 patients with locally-advanced or metastatic cutaneous squamous-cell carcinoma who were not eligible for surgery. The phase 2 component involved 59 patients with metastatic disease.

Patients were treated with intravenous cemiplimab every 2 weeks for 48 weeks in the phase 1 study, and up to 96 weeks – or until unacceptable toxicity or disease progression – in the phase 2 study.

In the phase 1 study, researchers saw a response rate of 50% and a 65% rate of durable disease control, after a median follow-up of 11 months (1.1-17). The median time to response was 2.3 months, and more than half the patients (54%) who showed a response maintained that response past 6 months.

In the phase 2 study in patients with metastatic disease, 47% responded to the treatment – 24 patients showed a partial response and 4 showed a complete response. Of those who responded, 61% showed durable disease control after a median follow-up of 7.9 months.

The median time to response in this group of patients was 1.9 months, and 57% of those who did respond still showed a response at 6 months. However neither median progression-free survival nor median overall survival had been reached at the point of data cut-off.

The treatment showed similar effects in patients with regional and distant metastatic disease.

Advanced cutaneous squamous-cell carcinoma was thought to be an ideal target for immunotherapy because the high mutation burden in the tumor meant it would be sensitive to effector T cell attack, wrote Michael R. Migden, MD, of the University of Texas MD Anderson Cancer Center, Houston, and his coauthors.

 

 


“In addition, the dramatically increased risk of cutaneous squamous-cell carcinoma among people with immunosuppression pointed to an important role for immune surveillance with this cancer,” the authors wrote.

In the phase 2 study, 29% of patients experienced a serious adverse event – including two cases of pneumonitis – and three patients (5%) discontinued treatment. There were three deaths due to adverse events: One patient died from pneumonia complications, one died in his sleep, and one patient died following hypercalcemia and deep vein thrombosis.

Aside from these, most adverse events were grade 1 or 2. Around one-quarter of patients experienced diarrhea (27%) or fatigue (24%), while the other most common adverse events were nausea (17%), constipation (15%) and rash (15%). The authors noted that these adverse events were similar to those seen in other PD-1 inhibitors.

“Our results are consistent with an emerging theme regarding the high efficacy of immune checkpoint blockade for the treatment of hypermutated cancers, since the mutation burden of cutaneous squamous-cell carcinoma is similar to that reported for advanced solid tumors with microsatellite instability,” the authors wrote.
 

 

Cemiplimab is now being tested in a phase 2 trial in patients with advanced basal cell carcinoma.

The study was supported by Regeneron Pharmaceuticals and Sanofi. Eight authors declared funding from Regeneron to conduct the trial. Ten authors were employees of Regeneron. Fifteen authors also declared funding and payments from pharmaceutical companies outside the submitted work. Four had nothing to disclose.

SOURCE: Migden M et al. NEJM, 2018; June 4. doi: 10.1056/NEJMoa1805131.

An immune checkpoint inhibitor that targets the PD-1 receptor has shown “robust” efficacy among patients with advanced cutaneous squamous-cell carcinoma, according to researchers.

A combined phase 1/phase 2 study, published in the New England Journal of Medicine and presented simultaneously at the annual meeting of the American Society of Clinical Oncology, looked at the effect of monoclonal antibody cemiplimab in an expansion cohort of 26 patients with locally-advanced or metastatic cutaneous squamous-cell carcinoma who were not eligible for surgery. The phase 2 component involved 59 patients with metastatic disease.

Patients were treated with intravenous cemiplimab every 2 weeks for 48 weeks in the phase 1 study, and up to 96 weeks – or until unacceptable toxicity or disease progression – in the phase 2 study.

In the phase 1 study, researchers saw a response rate of 50% and a 65% rate of durable disease control, after a median follow-up of 11 months (1.1-17). The median time to response was 2.3 months, and more than half the patients (54%) who showed a response maintained that response past 6 months.

In the phase 2 study in patients with metastatic disease, 47% responded to the treatment – 24 patients showed a partial response and 4 showed a complete response. Of those who responded, 61% showed durable disease control after a median follow-up of 7.9 months.

The median time to response in this group of patients was 1.9 months, and 57% of those who did respond still showed a response at 6 months. However neither median progression-free survival nor median overall survival had been reached at the point of data cut-off.

The treatment showed similar effects in patients with regional and distant metastatic disease.

Advanced cutaneous squamous-cell carcinoma was thought to be an ideal target for immunotherapy because the high mutation burden in the tumor meant it would be sensitive to effector T cell attack, wrote Michael R. Migden, MD, of the University of Texas MD Anderson Cancer Center, Houston, and his coauthors.

 

 


“In addition, the dramatically increased risk of cutaneous squamous-cell carcinoma among people with immunosuppression pointed to an important role for immune surveillance with this cancer,” the authors wrote.

In the phase 2 study, 29% of patients experienced a serious adverse event – including two cases of pneumonitis – and three patients (5%) discontinued treatment. There were three deaths due to adverse events: One patient died from pneumonia complications, one died in his sleep, and one patient died following hypercalcemia and deep vein thrombosis.

Aside from these, most adverse events were grade 1 or 2. Around one-quarter of patients experienced diarrhea (27%) or fatigue (24%), while the other most common adverse events were nausea (17%), constipation (15%) and rash (15%). The authors noted that these adverse events were similar to those seen in other PD-1 inhibitors.

“Our results are consistent with an emerging theme regarding the high efficacy of immune checkpoint blockade for the treatment of hypermutated cancers, since the mutation burden of cutaneous squamous-cell carcinoma is similar to that reported for advanced solid tumors with microsatellite instability,” the authors wrote.
 

 

Cemiplimab is now being tested in a phase 2 trial in patients with advanced basal cell carcinoma.

The study was supported by Regeneron Pharmaceuticals and Sanofi. Eight authors declared funding from Regeneron to conduct the trial. Ten authors were employees of Regeneron. Fifteen authors also declared funding and payments from pharmaceutical companies outside the submitted work. Four had nothing to disclose.

SOURCE: Migden M et al. NEJM, 2018; June 4. doi: 10.1056/NEJMoa1805131.

Publications
Publications
Topics
Article Type
Click for Credit Status
Ready
Sections
Article Source

FROM NEW ENGLAND JOURNAL OF MEDICINE

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Vitals

 

Key clinical point: PD-1 inhibitor cemiplimab shows significant response in advanced squamous-cell carcinoma.

Major finding: Around half of patients with advanced squamous-cell carcinoma responded to checkpoint inhibitor cemiplimab.

Study details: Phase 1 expanded cohort study of 26 patients with advanced cutaneous squamous-cell carcinoma and phase 2 study of 59 patients with metastatic squamous-cell carcinoma.

Disclosures: The study was supported by Regeneron Pharmaceuticals and Sanofi. Eight authors declared funding from Regeneron to conduct the trial. Ten authors were employees of Regeneron. Fifteen authors declared funding and payments from pharmaceutical companies outside the submitted work. Four had nothing to disclose.

Source: Migden M et al. N Engl J Med. 2018 June 4. doi: 10.1056/NEJMoa1805131.

Disqus Comments
Default
Use ProPublica

FDA alerts clinicians to gastric balloon deaths

Article Type
Changed
Fri, 01/18/2019 - 17:42

 

A total of 12 deaths over the past 2 years have been linked to the use of liquid-filled intragastric balloon devices for the treatment of obesity, according to an alert from the Food and Drug Administration issued on June 4.

Seven of these deaths occurred in patients in the United States; four involved the ORBERA Intragastric Balloon System, and three involved the ReShape Integrated Dual Balloon System.

Four of the deaths reported worldwide since 2016 occurred following gastric perforation within a month of surgery (three with the ORBERA system and one with the ReShape system), according to the FDA. A fifth death involving the Orbera system remains under investigation by the manufacturer.

The FDA has approved updated labeling for the ORBERA and ReShape balloon systems in the United States. The labels contain more information about possible death associated with the use of these devices in the United States. The manufacturers’ sites, Apollo Endosurgery and ReShape Lifesciences, provide more details about the new labeling.

In a letter to health care providers, the FDA advised clinicians to educate bariatric surgery patients about the symptoms of complications from balloon procedures, including not only gastric perforation but also esophageal perforation, balloon deflation, gastrointestinal obstruction, and ulceration. In addition, the FDA reminded clinicians to monitor patients during the entire course of treatment for additional complications, including acute pancreatitis and spontaneous hyperinflation.

Any adverse events involving intragastric balloon systems should be reported to the FDA through MedWatch, the FDA Safety Information and Adverse Event Reporting program.

Publications
Topics
Sections

 

A total of 12 deaths over the past 2 years have been linked to the use of liquid-filled intragastric balloon devices for the treatment of obesity, according to an alert from the Food and Drug Administration issued on June 4.

Seven of these deaths occurred in patients in the United States; four involved the ORBERA Intragastric Balloon System, and three involved the ReShape Integrated Dual Balloon System.

Four of the deaths reported worldwide since 2016 occurred following gastric perforation within a month of surgery (three with the ORBERA system and one with the ReShape system), according to the FDA. A fifth death involving the Orbera system remains under investigation by the manufacturer.

The FDA has approved updated labeling for the ORBERA and ReShape balloon systems in the United States. The labels contain more information about possible death associated with the use of these devices in the United States. The manufacturers’ sites, Apollo Endosurgery and ReShape Lifesciences, provide more details about the new labeling.

In a letter to health care providers, the FDA advised clinicians to educate bariatric surgery patients about the symptoms of complications from balloon procedures, including not only gastric perforation but also esophageal perforation, balloon deflation, gastrointestinal obstruction, and ulceration. In addition, the FDA reminded clinicians to monitor patients during the entire course of treatment for additional complications, including acute pancreatitis and spontaneous hyperinflation.

Any adverse events involving intragastric balloon systems should be reported to the FDA through MedWatch, the FDA Safety Information and Adverse Event Reporting program.

 

A total of 12 deaths over the past 2 years have been linked to the use of liquid-filled intragastric balloon devices for the treatment of obesity, according to an alert from the Food and Drug Administration issued on June 4.

Seven of these deaths occurred in patients in the United States; four involved the ORBERA Intragastric Balloon System, and three involved the ReShape Integrated Dual Balloon System.

Four of the deaths reported worldwide since 2016 occurred following gastric perforation within a month of surgery (three with the ORBERA system and one with the ReShape system), according to the FDA. A fifth death involving the Orbera system remains under investigation by the manufacturer.

The FDA has approved updated labeling for the ORBERA and ReShape balloon systems in the United States. The labels contain more information about possible death associated with the use of these devices in the United States. The manufacturers’ sites, Apollo Endosurgery and ReShape Lifesciences, provide more details about the new labeling.

In a letter to health care providers, the FDA advised clinicians to educate bariatric surgery patients about the symptoms of complications from balloon procedures, including not only gastric perforation but also esophageal perforation, balloon deflation, gastrointestinal obstruction, and ulceration. In addition, the FDA reminded clinicians to monitor patients during the entire course of treatment for additional complications, including acute pancreatitis and spontaneous hyperinflation.

Any adverse events involving intragastric balloon systems should be reported to the FDA through MedWatch, the FDA Safety Information and Adverse Event Reporting program.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica

MRD-negative status signals better outcomes in CAR T–treated ALL

Article Type
Changed
Fri, 01/04/2019 - 10:26

 

– Minimal residual disease (MRD)–negative complete remission was strongly associated with improved survival outcomes in patients with B-cell acute lymphocytic leukemia (ALL) who received CD19 chimeric antigen receptor (CAR) T cells, results of a retrospective study showed.

Allogeneic hematopoietic stem cell transplant (HSCT) appeared to improve both disease-free and overall survival in those patients who had achieved MRD-negative complete remission, according to results of the study, which were presented at the annual meeting of the American Society of Clinical Oncology.

“Based upon our interaction testing, the potential benefit [of transplant] appears to exist in both good-risk and bad-risk patients as identified through multivariate modeling,” said study investigator Kevin Anthony Hay, MD, of Fred Hutchinson Cancer Research Center, Seattle.

In a comment on the results, Sarah Cooley, MD, noted that the benefits of allogeneic transplant were apparent regardless of whether the patients met criteria for the good-risk subgroup, which was defined by levels of lactate dehydrogenase (LDH) and platelets along with exposure to fludarabine as part of the conditioning regimen.

“I think this suggests that the goal at this point is to get patients to an MRD-negative state and to potentially curative transplant,” said Dr. Cooley, director of investigator-initiated research at Masonic Medical Center at the University of Minnesota, Minneapolis.

The retrospective analysis by Dr. Hay and his colleagues included 53 adults with relapsed or refractory ALL who had bone marrow or extramedullary disease at baseline and had received CD19 CAR T cells at or under the maximum tolerated dose at least 1 year prior to this analysis. Of that group, 45 (85%) achieved MRD-negative complete remission.

Those patients who did achieve MRD-negative complete remission had an improved median disease-free survival at 7.6 months versus 0.8 months (P less than .0001) and improved overall survival at 20.0 months versus 5.0 months (P = 0.014).

 

 


Most of the MRD-negative patients who relapsed did so within the first 6 months, an observation that led investigators to consider whether factors exist that could predict better outcomes.

In a multivariate analysis, they found three variables associated with disease free survival: higher LDH prior to lymphodepletion (hazard ratio, 1.39), along with higher platelet count prior to lymphodepletion and incorporation of fludarabine into the regimen, with hazard ratios of 0.65 and 0.34, respectively.

Using those three characteristics, investigators grouped patients as “good risk” if they had normal LDH, platelet count at or above 100 prior to lymphodepletion that included fludarabine. The 24-month disease-free survival for good-risk patients was 78%, and overall survival was 86%.

The role of allogeneic HSCT after ALL patients achieved MRD-negative complete remission with CAR T-cell therapy was one of the “major questions in the field,” Dr. Hay said.
 

 


In this analysis, Dr. Hay and colleagues found that patients who underwent transplant in MRD-negative complete remission had a 24-month disease free survival and overall survival of 61% and 72%, respectively, both of which were significantly higher than in patients with MRD-negative complete remission who had no transplant.

The disease-free survival benefit was not specific to the good-risk group, according to Dr. Hay, who said an interaction test demonstrated no significant interaction between risk group and allogeneic HSCT after CAR T-cell infusion (P = 0.53).

“This is a very important finding that should be further [studied] in an appropriately designed clinical trial,” Dr. Hay said during an oral presentation of the study results.

Dr. Hay and several coauthors reported financial disclosures related to Juno Therapeutics. Other disclosures reported by study coauthors included Cell Medica, Celgene, Eureka Therapeutics, Genentech/Roche, Gilead Sciences, Kite Pharma, Novartis, and others.

SOURCE: Hay KA. ASCO 2018, Abstract 7005.

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

 

– Minimal residual disease (MRD)–negative complete remission was strongly associated with improved survival outcomes in patients with B-cell acute lymphocytic leukemia (ALL) who received CD19 chimeric antigen receptor (CAR) T cells, results of a retrospective study showed.

Allogeneic hematopoietic stem cell transplant (HSCT) appeared to improve both disease-free and overall survival in those patients who had achieved MRD-negative complete remission, according to results of the study, which were presented at the annual meeting of the American Society of Clinical Oncology.

“Based upon our interaction testing, the potential benefit [of transplant] appears to exist in both good-risk and bad-risk patients as identified through multivariate modeling,” said study investigator Kevin Anthony Hay, MD, of Fred Hutchinson Cancer Research Center, Seattle.

In a comment on the results, Sarah Cooley, MD, noted that the benefits of allogeneic transplant were apparent regardless of whether the patients met criteria for the good-risk subgroup, which was defined by levels of lactate dehydrogenase (LDH) and platelets along with exposure to fludarabine as part of the conditioning regimen.

“I think this suggests that the goal at this point is to get patients to an MRD-negative state and to potentially curative transplant,” said Dr. Cooley, director of investigator-initiated research at Masonic Medical Center at the University of Minnesota, Minneapolis.

The retrospective analysis by Dr. Hay and his colleagues included 53 adults with relapsed or refractory ALL who had bone marrow or extramedullary disease at baseline and had received CD19 CAR T cells at or under the maximum tolerated dose at least 1 year prior to this analysis. Of that group, 45 (85%) achieved MRD-negative complete remission.

Those patients who did achieve MRD-negative complete remission had an improved median disease-free survival at 7.6 months versus 0.8 months (P less than .0001) and improved overall survival at 20.0 months versus 5.0 months (P = 0.014).

 

 


Most of the MRD-negative patients who relapsed did so within the first 6 months, an observation that led investigators to consider whether factors exist that could predict better outcomes.

In a multivariate analysis, they found three variables associated with disease free survival: higher LDH prior to lymphodepletion (hazard ratio, 1.39), along with higher platelet count prior to lymphodepletion and incorporation of fludarabine into the regimen, with hazard ratios of 0.65 and 0.34, respectively.

Using those three characteristics, investigators grouped patients as “good risk” if they had normal LDH, platelet count at or above 100 prior to lymphodepletion that included fludarabine. The 24-month disease-free survival for good-risk patients was 78%, and overall survival was 86%.

The role of allogeneic HSCT after ALL patients achieved MRD-negative complete remission with CAR T-cell therapy was one of the “major questions in the field,” Dr. Hay said.
 

 


In this analysis, Dr. Hay and colleagues found that patients who underwent transplant in MRD-negative complete remission had a 24-month disease free survival and overall survival of 61% and 72%, respectively, both of which were significantly higher than in patients with MRD-negative complete remission who had no transplant.

The disease-free survival benefit was not specific to the good-risk group, according to Dr. Hay, who said an interaction test demonstrated no significant interaction between risk group and allogeneic HSCT after CAR T-cell infusion (P = 0.53).

“This is a very important finding that should be further [studied] in an appropriately designed clinical trial,” Dr. Hay said during an oral presentation of the study results.

Dr. Hay and several coauthors reported financial disclosures related to Juno Therapeutics. Other disclosures reported by study coauthors included Cell Medica, Celgene, Eureka Therapeutics, Genentech/Roche, Gilead Sciences, Kite Pharma, Novartis, and others.

SOURCE: Hay KA. ASCO 2018, Abstract 7005.

 

– Minimal residual disease (MRD)–negative complete remission was strongly associated with improved survival outcomes in patients with B-cell acute lymphocytic leukemia (ALL) who received CD19 chimeric antigen receptor (CAR) T cells, results of a retrospective study showed.

Allogeneic hematopoietic stem cell transplant (HSCT) appeared to improve both disease-free and overall survival in those patients who had achieved MRD-negative complete remission, according to results of the study, which were presented at the annual meeting of the American Society of Clinical Oncology.

“Based upon our interaction testing, the potential benefit [of transplant] appears to exist in both good-risk and bad-risk patients as identified through multivariate modeling,” said study investigator Kevin Anthony Hay, MD, of Fred Hutchinson Cancer Research Center, Seattle.

In a comment on the results, Sarah Cooley, MD, noted that the benefits of allogeneic transplant were apparent regardless of whether the patients met criteria for the good-risk subgroup, which was defined by levels of lactate dehydrogenase (LDH) and platelets along with exposure to fludarabine as part of the conditioning regimen.

“I think this suggests that the goal at this point is to get patients to an MRD-negative state and to potentially curative transplant,” said Dr. Cooley, director of investigator-initiated research at Masonic Medical Center at the University of Minnesota, Minneapolis.

The retrospective analysis by Dr. Hay and his colleagues included 53 adults with relapsed or refractory ALL who had bone marrow or extramedullary disease at baseline and had received CD19 CAR T cells at or under the maximum tolerated dose at least 1 year prior to this analysis. Of that group, 45 (85%) achieved MRD-negative complete remission.

Those patients who did achieve MRD-negative complete remission had an improved median disease-free survival at 7.6 months versus 0.8 months (P less than .0001) and improved overall survival at 20.0 months versus 5.0 months (P = 0.014).

 

 


Most of the MRD-negative patients who relapsed did so within the first 6 months, an observation that led investigators to consider whether factors exist that could predict better outcomes.

In a multivariate analysis, they found three variables associated with disease free survival: higher LDH prior to lymphodepletion (hazard ratio, 1.39), along with higher platelet count prior to lymphodepletion and incorporation of fludarabine into the regimen, with hazard ratios of 0.65 and 0.34, respectively.

Using those three characteristics, investigators grouped patients as “good risk” if they had normal LDH, platelet count at or above 100 prior to lymphodepletion that included fludarabine. The 24-month disease-free survival for good-risk patients was 78%, and overall survival was 86%.

The role of allogeneic HSCT after ALL patients achieved MRD-negative complete remission with CAR T-cell therapy was one of the “major questions in the field,” Dr. Hay said.
 

 


In this analysis, Dr. Hay and colleagues found that patients who underwent transplant in MRD-negative complete remission had a 24-month disease free survival and overall survival of 61% and 72%, respectively, both of which were significantly higher than in patients with MRD-negative complete remission who had no transplant.

The disease-free survival benefit was not specific to the good-risk group, according to Dr. Hay, who said an interaction test demonstrated no significant interaction between risk group and allogeneic HSCT after CAR T-cell infusion (P = 0.53).

“This is a very important finding that should be further [studied] in an appropriately designed clinical trial,” Dr. Hay said during an oral presentation of the study results.

Dr. Hay and several coauthors reported financial disclosures related to Juno Therapeutics. Other disclosures reported by study coauthors included Cell Medica, Celgene, Eureka Therapeutics, Genentech/Roche, Gilead Sciences, Kite Pharma, Novartis, and others.

SOURCE: Hay KA. ASCO 2018, Abstract 7005.

Publications
Publications
Topics
Article Type
Sections
Article Source

REPORTING FROM ASCO 2018

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Vitals

 

Key clinical point: Minimal residual disease (MRD)–negative complete remission was strongly associated with improved survival outcomes.

Major finding: Patients who achieved MRD-negative complete remission had an improved median disease-free survival at 7.6 months versus 0.8 months (P less than .0001)

Study details: A retrospective analysis including 53 patients with ALL who had bone marrow or extramedullary disease at baseline and had received CD19 CAR T cells at or under the maximum tolerated dose at least 1 year prior to this analysis.

Disclosures: Researchers reported financial ties to Juno Therapeutics, Cell Medica, Celgene, Eureka Therapeutics, Genentech/Roche, Gilead Sciences, Kite Pharma, Novartis, and others.

Source: Hay KA. ASCO 2018, Abstract 7005.

Disqus Comments
Default
Use ProPublica

Dr. William J. Gradishar shares breast cancer take-aways from ASCO 2018

Article Type
Changed
Wed, 01/04/2023 - 16:45

– William J. Gradishar, MD, discussed the clinical impact of breast cancer research presented at the annual meeting of the American Society of Clinical Oncology.

In a video interview, Dr. Gradishar, the Betsy Bramsen Professor of Breast Oncology at Northwestern University, Chicago, said TAILORx was a “big win” in that it has no doubt diminished the number of women with early-stage breast cancer who will require chemotherapy. However, although the trial has provided some clarity, it also has left some questions open, particularly for patients under 50 years of age, he said.

Dr. Gradishar also discussed the results of combination trials of targeted therapy with either endocrine therapy or chemotherapy. In discussing SANDPIPER, which evaluated whether a phosphoinositide 3-kinase inhibitor could enhance the effect of anti-hormonal therapy, he said that although it was a positive trial, “from a clinician’s standpoint, it’s probably not sufficient in my mind to get really excited about.”

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

– William J. Gradishar, MD, discussed the clinical impact of breast cancer research presented at the annual meeting of the American Society of Clinical Oncology.

In a video interview, Dr. Gradishar, the Betsy Bramsen Professor of Breast Oncology at Northwestern University, Chicago, said TAILORx was a “big win” in that it has no doubt diminished the number of women with early-stage breast cancer who will require chemotherapy. However, although the trial has provided some clarity, it also has left some questions open, particularly for patients under 50 years of age, he said.

Dr. Gradishar also discussed the results of combination trials of targeted therapy with either endocrine therapy or chemotherapy. In discussing SANDPIPER, which evaluated whether a phosphoinositide 3-kinase inhibitor could enhance the effect of anti-hormonal therapy, he said that although it was a positive trial, “from a clinician’s standpoint, it’s probably not sufficient in my mind to get really excited about.”

– William J. Gradishar, MD, discussed the clinical impact of breast cancer research presented at the annual meeting of the American Society of Clinical Oncology.

In a video interview, Dr. Gradishar, the Betsy Bramsen Professor of Breast Oncology at Northwestern University, Chicago, said TAILORx was a “big win” in that it has no doubt diminished the number of women with early-stage breast cancer who will require chemotherapy. However, although the trial has provided some clarity, it also has left some questions open, particularly for patients under 50 years of age, he said.

Dr. Gradishar also discussed the results of combination trials of targeted therapy with either endocrine therapy or chemotherapy. In discussing SANDPIPER, which evaluated whether a phosphoinositide 3-kinase inhibitor could enhance the effect of anti-hormonal therapy, he said that although it was a positive trial, “from a clinician’s standpoint, it’s probably not sufficient in my mind to get really excited about.”

Publications
Publications
Topics
Article Type
Sections
Article Source

REPORTING FROM ASCO 2018

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

Beltlike Lichen Planus Pigmentosus Complicated With Focal Amyloidosis

Article Type
Changed
Thu, 01/10/2019 - 13:51
Display Headline
Beltlike Lichen Planus Pigmentosus Complicated With Focal Amyloidosis

To the Editor:

A 68-year-old man presented with slightly itchy macules on the waist and abdomen of approximately 2 years’ duration. He reported that the initial lesions were dark red and subsequently coalesced to form a beltlike pigmentation on the abdomen. He denied any prior treatment, and the lesions did not spontaneously resolve. The patient was taking escitalopram oxalate, telmisartan, and aspirin for depression and cardiovascular disease that was diagnosed 3 years prior. He reported no exposure to UV radiation or a heat source. He denied use of any cosmetics on the body as well as a family history of similar symptoms.

Physical examination showed reticulate brown-purple macules with slight scale on the surface that had become confluent, forming a beltlike pigmentation on the waist and abdomen (Figure 1). Wickham striae were not seen. The oral mucosa and nails were not affected. Microscopic examination for fungal infections was negative.

Figure1
Figure 1. Reticulate brown-purple macules with slight scale on the surface that had become confluent, forming a beltlike pigmentation on the waist and abdomen.

Systematic physical and laboratory examinations revealed no abnormalities. A skin biopsy from a macule on the abdomen showed hyperkeratosis, thinned out stratum spinosum with flattening of rete ridges, hypergranulosis with vacuolar alteration of the basal cell layer, and bandlike infiltration of lymphocytes and melanophages with incontinence of pigment (Figure 2). Focalized purplish homogeneous deposits were observed in the upper dermis (Figure 3), of which positive crystal violet staining indicated amyloidosis (Figure 4). Congo red stain revealed amyloid deposition (Figure 5). Thus, the diagnosis of lichen planus pigmentosus (LPP) complicated with focal amyloidosis was made. The patient was treated with topical corticosteroids and tretinoin, and no notable therapeutic effects were observed at 3-month follow-up.

Figure2
Figure 2. Histopathology showed hyperkeratosis, thinned out stratum spinosum with flattening of rete ridges, hypergranulosis with vacuolar alteration of the basal cell layer, and bandlike infiltration of lymphocytes and melanophages with incontinence of pigment (H&E, original magnification ×10).

Figure3
Figure 3. Focalized purplish homogeneous deposits were observed in the upper dermis (H&E, original magnification ×40).

Figure4
Figure 4. Crystal violet staining was positive for amyloidosis (original magnification ×40).

Figure5
Figure 5. Congo red stain revealed amyloid deposition (original magnification ×40).

Lichen planus pigmentosus, a variant of lichen planus, is a condition of unknown etiology exhibiting dark brown macules and/or papules and a long clinical course. The face, neck, trunk, arms, and legs are the most common areas of presentation, whereas involvement of the scalp, nails, or oral mucosa is relatively rare.

The first clinicohistopathological study with a large sample size was documented by Bhutani et al1 in 1974 who termed the currently recognized entity lichen planus pigmentosus. Lichen planus pigmentosus is a frequently encountered hyperpigmentation disorder in Indians, whereas sporadic cases also are reported in other regions and ethnicities.2 In cases of LPP, the pigmentation is symmetrical, and its pattern most often is diffuse, then reticular, blotchy, and perifollicular.3 Two unique patterns of LPP have been documented, including linear/blaschkoid LPP and zosteriform LPP.4,5 Our patient showed a unique beltlike distribution pattern.

The pathogenesis of LPP still is unclear, and several inciting factors such as mustard oil, gold therapy,6 and hepatitis C virus infection have been cited.7 Mancuso and Berdondini8 reported a case of LPP flaring immediately after relapse of nephrotic syndrome. It also has been considered as a paraneoplastic phenomen.9 No exact cause was found in our patient after a series of relative examinations.

The histopathologic changes associated with LPP consist of atrophic epidermis; bandlike lymphocytic infiltrate with vacuolar degeneration of the basal layer in the epidermis; and prominent melanin incontinence in the upper dermis, which can be diverse depending on different sites of skin biopsy and the phase of LPP. Histopathologic findings in our patient were consistent with LPP. The differential diagnosis for the reticulate pattern of pigmentation seen in our patient included confluent and reticulated papillomatosis and poikilodermalike cutaneous amyloidosis, both easily excluded with histopathologic confirmation.

Local amyloidosis also was confirmed by crystal violet staining in our case and its etiology was uncertain. Generalized and local amyloidosis has been reported in association with lichen planus. The diagnosis of lichen planus was followed by the diagnosis of amyloidosis, and the typical skin lesions of these 2 conditions were able to be differentiated in these reported cases.10,11 However, beltlike pigmentation was the only manifestation for our patient and we could not separate the 2 conditions with the naked eye.

Chronic irritation to the skin resulting in excessive production of degenerate keratins and their subsequent conversion into amyloid deposits has been proposed to be an etiologic factor of amyloidosis.11 Because of the distribution pattern in our case, we believe focal amyloidosis could be attributed to chronic friction and scratching.

References
  1. Bhutani LK, Bedi TR, Pandhi RK, et al. Lichen planus pigmentosus. Dermatologica. 1974;149:43-50.
  2. Kanwar AJ, Kaur S. Lichen planus pigmentosus. J Am Acad Dermatol. 1989;21(4, pt 1):815.
  3. Kanwar AJ, Dogra S, Handa S, et al. A study of 124 Indian patients with lichen planus pigmentosus. Clin Exp Dermatol. 2003;28:481-485.
  4. Akarsu S, Ilknur T, Özer E, et al. Lichen planus pigmentosus distributed along the lines of Blaschko. Int J Dermatol. 2013;52:253-254.
  5. Cho S, Whang KK. Lichen planus pigmentosus presenting in zosteriform pattern. J Dermatol. 1997;24:193-197.
  6. Ingber A, Weissmann-Katzenelson V, David M, et al. Lichen planus and lichen planus pigmentosus following gold therapy—case reports and review of the literature [in German]. Z Hautkr. 1986;61:315-319.
  7. Al-Mutairi N, El-Khalawany M. Clinicopathological characteristics of lichen planus pigmentosus and its response to tacrolimus ointment: an open label, non-randomized, prospective study. J Eur Acad Dermatol Venereol. 2010;24:535-540.
  8. Mancuso G, Berdondini RM. Coexistence of lichen planus pigmentosus and minimal change nephrotic syndrome. Eur J Dermatol. 2009;19:389-390.
  9. Sassolas B, Zagnoli A, Leroy JP, et al. Lichen planus pigmentosus associated with acrokeratosis of Bazex. Clin Exp Dermatol. 1994;19:70-73.
  10. Maeda H, Ohta S, Saito Y, et al. Epidermal origin of the amyloid in localized cutaneous amyloidosis. Br J Dermatol. 1982;106:345-351.
  11. Hongcharu W, Baldassano M, Gonzalez E. Generalized lichen amyloidosis associated with chronic lichen planus. J Am Acad Dermatol. 2000;43:346-348.
Article PDF
Author and Disclosure Information

From the Department of Dermatology, First Affiliated Hospital, Nanjing Medical University, China.

The authors report no conflict of interest.

Correspondence: Yan Lu, MD, PhD, Department of Dermatology, First Affiliated Hospital, Nanjing Medical University, #300 Guangzhou Rd, Nanjing, Jiangsu Province 210029, China ([email protected]).

Issue
Cutis - 101(5)
Publications
Topics
Page Number
E24-E26
Sections
Author and Disclosure Information

From the Department of Dermatology, First Affiliated Hospital, Nanjing Medical University, China.

The authors report no conflict of interest.

Correspondence: Yan Lu, MD, PhD, Department of Dermatology, First Affiliated Hospital, Nanjing Medical University, #300 Guangzhou Rd, Nanjing, Jiangsu Province 210029, China ([email protected]).

Author and Disclosure Information

From the Department of Dermatology, First Affiliated Hospital, Nanjing Medical University, China.

The authors report no conflict of interest.

Correspondence: Yan Lu, MD, PhD, Department of Dermatology, First Affiliated Hospital, Nanjing Medical University, #300 Guangzhou Rd, Nanjing, Jiangsu Province 210029, China ([email protected]).

Article PDF
Article PDF

To the Editor:

A 68-year-old man presented with slightly itchy macules on the waist and abdomen of approximately 2 years’ duration. He reported that the initial lesions were dark red and subsequently coalesced to form a beltlike pigmentation on the abdomen. He denied any prior treatment, and the lesions did not spontaneously resolve. The patient was taking escitalopram oxalate, telmisartan, and aspirin for depression and cardiovascular disease that was diagnosed 3 years prior. He reported no exposure to UV radiation or a heat source. He denied use of any cosmetics on the body as well as a family history of similar symptoms.

Physical examination showed reticulate brown-purple macules with slight scale on the surface that had become confluent, forming a beltlike pigmentation on the waist and abdomen (Figure 1). Wickham striae were not seen. The oral mucosa and nails were not affected. Microscopic examination for fungal infections was negative.

Figure1
Figure 1. Reticulate brown-purple macules with slight scale on the surface that had become confluent, forming a beltlike pigmentation on the waist and abdomen.

Systematic physical and laboratory examinations revealed no abnormalities. A skin biopsy from a macule on the abdomen showed hyperkeratosis, thinned out stratum spinosum with flattening of rete ridges, hypergranulosis with vacuolar alteration of the basal cell layer, and bandlike infiltration of lymphocytes and melanophages with incontinence of pigment (Figure 2). Focalized purplish homogeneous deposits were observed in the upper dermis (Figure 3), of which positive crystal violet staining indicated amyloidosis (Figure 4). Congo red stain revealed amyloid deposition (Figure 5). Thus, the diagnosis of lichen planus pigmentosus (LPP) complicated with focal amyloidosis was made. The patient was treated with topical corticosteroids and tretinoin, and no notable therapeutic effects were observed at 3-month follow-up.

Figure2
Figure 2. Histopathology showed hyperkeratosis, thinned out stratum spinosum with flattening of rete ridges, hypergranulosis with vacuolar alteration of the basal cell layer, and bandlike infiltration of lymphocytes and melanophages with incontinence of pigment (H&E, original magnification ×10).

Figure3
Figure 3. Focalized purplish homogeneous deposits were observed in the upper dermis (H&E, original magnification ×40).

Figure4
Figure 4. Crystal violet staining was positive for amyloidosis (original magnification ×40).

Figure5
Figure 5. Congo red stain revealed amyloid deposition (original magnification ×40).

Lichen planus pigmentosus, a variant of lichen planus, is a condition of unknown etiology exhibiting dark brown macules and/or papules and a long clinical course. The face, neck, trunk, arms, and legs are the most common areas of presentation, whereas involvement of the scalp, nails, or oral mucosa is relatively rare.

The first clinicohistopathological study with a large sample size was documented by Bhutani et al1 in 1974 who termed the currently recognized entity lichen planus pigmentosus. Lichen planus pigmentosus is a frequently encountered hyperpigmentation disorder in Indians, whereas sporadic cases also are reported in other regions and ethnicities.2 In cases of LPP, the pigmentation is symmetrical, and its pattern most often is diffuse, then reticular, blotchy, and perifollicular.3 Two unique patterns of LPP have been documented, including linear/blaschkoid LPP and zosteriform LPP.4,5 Our patient showed a unique beltlike distribution pattern.

The pathogenesis of LPP still is unclear, and several inciting factors such as mustard oil, gold therapy,6 and hepatitis C virus infection have been cited.7 Mancuso and Berdondini8 reported a case of LPP flaring immediately after relapse of nephrotic syndrome. It also has been considered as a paraneoplastic phenomen.9 No exact cause was found in our patient after a series of relative examinations.

The histopathologic changes associated with LPP consist of atrophic epidermis; bandlike lymphocytic infiltrate with vacuolar degeneration of the basal layer in the epidermis; and prominent melanin incontinence in the upper dermis, which can be diverse depending on different sites of skin biopsy and the phase of LPP. Histopathologic findings in our patient were consistent with LPP. The differential diagnosis for the reticulate pattern of pigmentation seen in our patient included confluent and reticulated papillomatosis and poikilodermalike cutaneous amyloidosis, both easily excluded with histopathologic confirmation.

Local amyloidosis also was confirmed by crystal violet staining in our case and its etiology was uncertain. Generalized and local amyloidosis has been reported in association with lichen planus. The diagnosis of lichen planus was followed by the diagnosis of amyloidosis, and the typical skin lesions of these 2 conditions were able to be differentiated in these reported cases.10,11 However, beltlike pigmentation was the only manifestation for our patient and we could not separate the 2 conditions with the naked eye.

Chronic irritation to the skin resulting in excessive production of degenerate keratins and their subsequent conversion into amyloid deposits has been proposed to be an etiologic factor of amyloidosis.11 Because of the distribution pattern in our case, we believe focal amyloidosis could be attributed to chronic friction and scratching.

To the Editor:

A 68-year-old man presented with slightly itchy macules on the waist and abdomen of approximately 2 years’ duration. He reported that the initial lesions were dark red and subsequently coalesced to form a beltlike pigmentation on the abdomen. He denied any prior treatment, and the lesions did not spontaneously resolve. The patient was taking escitalopram oxalate, telmisartan, and aspirin for depression and cardiovascular disease that was diagnosed 3 years prior. He reported no exposure to UV radiation or a heat source. He denied use of any cosmetics on the body as well as a family history of similar symptoms.

Physical examination showed reticulate brown-purple macules with slight scale on the surface that had become confluent, forming a beltlike pigmentation on the waist and abdomen (Figure 1). Wickham striae were not seen. The oral mucosa and nails were not affected. Microscopic examination for fungal infections was negative.

Figure1
Figure 1. Reticulate brown-purple macules with slight scale on the surface that had become confluent, forming a beltlike pigmentation on the waist and abdomen.

Systematic physical and laboratory examinations revealed no abnormalities. A skin biopsy from a macule on the abdomen showed hyperkeratosis, thinned out stratum spinosum with flattening of rete ridges, hypergranulosis with vacuolar alteration of the basal cell layer, and bandlike infiltration of lymphocytes and melanophages with incontinence of pigment (Figure 2). Focalized purplish homogeneous deposits were observed in the upper dermis (Figure 3), of which positive crystal violet staining indicated amyloidosis (Figure 4). Congo red stain revealed amyloid deposition (Figure 5). Thus, the diagnosis of lichen planus pigmentosus (LPP) complicated with focal amyloidosis was made. The patient was treated with topical corticosteroids and tretinoin, and no notable therapeutic effects were observed at 3-month follow-up.

Figure2
Figure 2. Histopathology showed hyperkeratosis, thinned out stratum spinosum with flattening of rete ridges, hypergranulosis with vacuolar alteration of the basal cell layer, and bandlike infiltration of lymphocytes and melanophages with incontinence of pigment (H&E, original magnification ×10).

Figure3
Figure 3. Focalized purplish homogeneous deposits were observed in the upper dermis (H&E, original magnification ×40).

Figure4
Figure 4. Crystal violet staining was positive for amyloidosis (original magnification ×40).

Figure5
Figure 5. Congo red stain revealed amyloid deposition (original magnification ×40).

Lichen planus pigmentosus, a variant of lichen planus, is a condition of unknown etiology exhibiting dark brown macules and/or papules and a long clinical course. The face, neck, trunk, arms, and legs are the most common areas of presentation, whereas involvement of the scalp, nails, or oral mucosa is relatively rare.

The first clinicohistopathological study with a large sample size was documented by Bhutani et al1 in 1974 who termed the currently recognized entity lichen planus pigmentosus. Lichen planus pigmentosus is a frequently encountered hyperpigmentation disorder in Indians, whereas sporadic cases also are reported in other regions and ethnicities.2 In cases of LPP, the pigmentation is symmetrical, and its pattern most often is diffuse, then reticular, blotchy, and perifollicular.3 Two unique patterns of LPP have been documented, including linear/blaschkoid LPP and zosteriform LPP.4,5 Our patient showed a unique beltlike distribution pattern.

The pathogenesis of LPP still is unclear, and several inciting factors such as mustard oil, gold therapy,6 and hepatitis C virus infection have been cited.7 Mancuso and Berdondini8 reported a case of LPP flaring immediately after relapse of nephrotic syndrome. It also has been considered as a paraneoplastic phenomen.9 No exact cause was found in our patient after a series of relative examinations.

The histopathologic changes associated with LPP consist of atrophic epidermis; bandlike lymphocytic infiltrate with vacuolar degeneration of the basal layer in the epidermis; and prominent melanin incontinence in the upper dermis, which can be diverse depending on different sites of skin biopsy and the phase of LPP. Histopathologic findings in our patient were consistent with LPP. The differential diagnosis for the reticulate pattern of pigmentation seen in our patient included confluent and reticulated papillomatosis and poikilodermalike cutaneous amyloidosis, both easily excluded with histopathologic confirmation.

Local amyloidosis also was confirmed by crystal violet staining in our case and its etiology was uncertain. Generalized and local amyloidosis has been reported in association with lichen planus. The diagnosis of lichen planus was followed by the diagnosis of amyloidosis, and the typical skin lesions of these 2 conditions were able to be differentiated in these reported cases.10,11 However, beltlike pigmentation was the only manifestation for our patient and we could not separate the 2 conditions with the naked eye.

Chronic irritation to the skin resulting in excessive production of degenerate keratins and their subsequent conversion into amyloid deposits has been proposed to be an etiologic factor of amyloidosis.11 Because of the distribution pattern in our case, we believe focal amyloidosis could be attributed to chronic friction and scratching.

References
  1. Bhutani LK, Bedi TR, Pandhi RK, et al. Lichen planus pigmentosus. Dermatologica. 1974;149:43-50.
  2. Kanwar AJ, Kaur S. Lichen planus pigmentosus. J Am Acad Dermatol. 1989;21(4, pt 1):815.
  3. Kanwar AJ, Dogra S, Handa S, et al. A study of 124 Indian patients with lichen planus pigmentosus. Clin Exp Dermatol. 2003;28:481-485.
  4. Akarsu S, Ilknur T, Özer E, et al. Lichen planus pigmentosus distributed along the lines of Blaschko. Int J Dermatol. 2013;52:253-254.
  5. Cho S, Whang KK. Lichen planus pigmentosus presenting in zosteriform pattern. J Dermatol. 1997;24:193-197.
  6. Ingber A, Weissmann-Katzenelson V, David M, et al. Lichen planus and lichen planus pigmentosus following gold therapy—case reports and review of the literature [in German]. Z Hautkr. 1986;61:315-319.
  7. Al-Mutairi N, El-Khalawany M. Clinicopathological characteristics of lichen planus pigmentosus and its response to tacrolimus ointment: an open label, non-randomized, prospective study. J Eur Acad Dermatol Venereol. 2010;24:535-540.
  8. Mancuso G, Berdondini RM. Coexistence of lichen planus pigmentosus and minimal change nephrotic syndrome. Eur J Dermatol. 2009;19:389-390.
  9. Sassolas B, Zagnoli A, Leroy JP, et al. Lichen planus pigmentosus associated with acrokeratosis of Bazex. Clin Exp Dermatol. 1994;19:70-73.
  10. Maeda H, Ohta S, Saito Y, et al. Epidermal origin of the amyloid in localized cutaneous amyloidosis. Br J Dermatol. 1982;106:345-351.
  11. Hongcharu W, Baldassano M, Gonzalez E. Generalized lichen amyloidosis associated with chronic lichen planus. J Am Acad Dermatol. 2000;43:346-348.
References
  1. Bhutani LK, Bedi TR, Pandhi RK, et al. Lichen planus pigmentosus. Dermatologica. 1974;149:43-50.
  2. Kanwar AJ, Kaur S. Lichen planus pigmentosus. J Am Acad Dermatol. 1989;21(4, pt 1):815.
  3. Kanwar AJ, Dogra S, Handa S, et al. A study of 124 Indian patients with lichen planus pigmentosus. Clin Exp Dermatol. 2003;28:481-485.
  4. Akarsu S, Ilknur T, Özer E, et al. Lichen planus pigmentosus distributed along the lines of Blaschko. Int J Dermatol. 2013;52:253-254.
  5. Cho S, Whang KK. Lichen planus pigmentosus presenting in zosteriform pattern. J Dermatol. 1997;24:193-197.
  6. Ingber A, Weissmann-Katzenelson V, David M, et al. Lichen planus and lichen planus pigmentosus following gold therapy—case reports and review of the literature [in German]. Z Hautkr. 1986;61:315-319.
  7. Al-Mutairi N, El-Khalawany M. Clinicopathological characteristics of lichen planus pigmentosus and its response to tacrolimus ointment: an open label, non-randomized, prospective study. J Eur Acad Dermatol Venereol. 2010;24:535-540.
  8. Mancuso G, Berdondini RM. Coexistence of lichen planus pigmentosus and minimal change nephrotic syndrome. Eur J Dermatol. 2009;19:389-390.
  9. Sassolas B, Zagnoli A, Leroy JP, et al. Lichen planus pigmentosus associated with acrokeratosis of Bazex. Clin Exp Dermatol. 1994;19:70-73.
  10. Maeda H, Ohta S, Saito Y, et al. Epidermal origin of the amyloid in localized cutaneous amyloidosis. Br J Dermatol. 1982;106:345-351.
  11. Hongcharu W, Baldassano M, Gonzalez E. Generalized lichen amyloidosis associated with chronic lichen planus. J Am Acad Dermatol. 2000;43:346-348.
Issue
Cutis - 101(5)
Issue
Cutis - 101(5)
Page Number
E24-E26
Page Number
E24-E26
Publications
Publications
Topics
Article Type
Display Headline
Beltlike Lichen Planus Pigmentosus Complicated With Focal Amyloidosis
Display Headline
Beltlike Lichen Planus Pigmentosus Complicated With Focal Amyloidosis
Sections
Inside the Article

Practice Points

  • Lichen planus pigmentosus can present in a unique beltlike distribution pattern.
  • Focal amyloidosis due to chronic friction and scratching cannot be excluded from the differential diagnosis.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Article PDF Media

Metastatic lung cancer: Pembrolizumab plus chemo prolongs survival

First-line pembrolizumab should be standard
Article Type
Changed
Fri, 01/04/2019 - 14:19

 

In patients with advanced non–small-cell lung cancer (NSCLC) without targetable mutations, the addition of pembrolizumab to first-line combination chemotherapy improves overall and progression-free survival, according to results from the phase 3 KEYNOTE-189 trial.

The researchers found that while patients in all subgroups benefited from the addition of pembrolizumab to chemotherapy, patients with a tumor proportion score of at least 50% benefited the most.

In 2017, accelerated approval was granted for pembrolizumab in combination with carboplatin and pemetrexed as first-line therapy for patients with NSCLC lacking EGFR and ALK mutations. This approval was based on response rates and progression-free survival from the KEYNOTE-024 phase 2 trial.

KEYNOTE-189 solidifies these findings, according to investigators. “Together with the results from KEYNOTE-024, the data from KEYNOTE-189 suggest that introducing immunotherapy as a first-line therapy may have a favorable long-term effect on outcomes,” wrote lead author Leena Gandhi, MD, director of the Perlmutter Cancer Center at New York University, and her colleagues. The report was published in the New England Journal of Medicine.

The double-blind, phase 3 trial assessed 616 treatment-naive patients with metastatic nonsquamous NSCLC without ALK or EGFR mutations. Patients received a platinum-based drug and pemetrexed plus placebo or 200 mg of pembrolizumab every 3 weeks for four cycles. After this, patients received placebo or pembrolizumab with pemetrexed maintenance therapy for up to 35 cycles. Primary endpoints were overall and progression-free survival, determined by a blinded radiologist.

At 12 months, the estimated overall survival was 69.2% for patients treated with pembrolizumab plus chemotherapy, compared with 49.4% for patients treated with chemotherapy alone. For patients treated with the pembrolizumab combination, median progression-free survival was 8.8 months, compared with 4.9 months for those treated with just chemotherapy. Median follow-up time was 10.5 months.

“The survival benefit associated with the pembrolizumab combination was observed in all subgroups of PD-L1 tumor proportion scores,” the authors wrote, “including patients with a score of less than 1%, a population for which single-agent PD-1 and PD-L1 inhibition have a small chance of benefit.” As with previous studies, the subgroup of patients with a tumor proportion score greater than or equal to 50% received the greatest benefit when pembrolizumab was added.


The authors noted that further research is needed to determine whether patients with high PD-L1 expression (TMS ≥ 50%) would benefit more from pembrolizumab combination therapy compared with pembrolizumab monotherapy, which is currently indicated.

SOURCE: Gandhi et al. N Engl J Med. 2018 May 31. doi: 10.1056/NEJMoa1801005.

Body

 

The recent phase 3 KEYNOTE-189 trial by Gandhi and her colleagues solidifies a shift in first-line standard therapy for some lung cancer patients, according to Joan H. Schiller, MD.

“This trial illustrates that PD-1 pathway inhibitors can be successfully combined with chemotherapy,” Dr. Schiller wrote in an editorial in the New England Journal of Medicine.

In the study, patients treated with carboplatin-pemetrexed-pembrolizumab therapy had longer overall and progression-free survival compared with patients treated with just chemotherapy.

“The magnitude of benefit is impressive,” Dr. Schiller wrote, “with a hazard ratio for death of 0.49 and 12-month overall survival of 69.2% in the pembrolizumab-combination group.”

However, “many unanswered questions remain,” Dr. Schiller said, particularly concerning the utility of the commonly used PD-L1 tumor proportion score, which “appears to be problematic, with some randomized trials showing benefit only with a score of 50% or more and others showing benefit at other cutoff points, including less than 1%.”

In the current study, all subgroups benefited from the addition of pembrolizumab, including patients with a score of less than 1%.

Despite the need for more research, Dr. Schiller said she believes that existing findings have revealed unprecedented benefits. “Is carboplatin-pemetrexed-pembrolizumab now the first-line standard therapy for patients with nonsquamous NSCLC lacking targetable mutations? In my opinion, the answer is yes.”

Dr. Schiller is with the Inova Schar Cancer Institute. These comments are adapted from an editorial (N Eng J Med. 2018 May 31. doi: 10.1056/NEJMe1804364 ). The author reported financial support from Merck, Lily, AstraZeneca, Genentech/Roche, OncoGenex, Halozyme, Synta, Vertex, Bristol-Myers Squibb, Clovis, Xcovery, AbbVie, Astex, Janssen, and Free to Breathe.

Publications
Topics
Sections
Body

 

The recent phase 3 KEYNOTE-189 trial by Gandhi and her colleagues solidifies a shift in first-line standard therapy for some lung cancer patients, according to Joan H. Schiller, MD.

“This trial illustrates that PD-1 pathway inhibitors can be successfully combined with chemotherapy,” Dr. Schiller wrote in an editorial in the New England Journal of Medicine.

In the study, patients treated with carboplatin-pemetrexed-pembrolizumab therapy had longer overall and progression-free survival compared with patients treated with just chemotherapy.

“The magnitude of benefit is impressive,” Dr. Schiller wrote, “with a hazard ratio for death of 0.49 and 12-month overall survival of 69.2% in the pembrolizumab-combination group.”

However, “many unanswered questions remain,” Dr. Schiller said, particularly concerning the utility of the commonly used PD-L1 tumor proportion score, which “appears to be problematic, with some randomized trials showing benefit only with a score of 50% or more and others showing benefit at other cutoff points, including less than 1%.”

In the current study, all subgroups benefited from the addition of pembrolizumab, including patients with a score of less than 1%.

Despite the need for more research, Dr. Schiller said she believes that existing findings have revealed unprecedented benefits. “Is carboplatin-pemetrexed-pembrolizumab now the first-line standard therapy for patients with nonsquamous NSCLC lacking targetable mutations? In my opinion, the answer is yes.”

Dr. Schiller is with the Inova Schar Cancer Institute. These comments are adapted from an editorial (N Eng J Med. 2018 May 31. doi: 10.1056/NEJMe1804364 ). The author reported financial support from Merck, Lily, AstraZeneca, Genentech/Roche, OncoGenex, Halozyme, Synta, Vertex, Bristol-Myers Squibb, Clovis, Xcovery, AbbVie, Astex, Janssen, and Free to Breathe.

Body

 

The recent phase 3 KEYNOTE-189 trial by Gandhi and her colleagues solidifies a shift in first-line standard therapy for some lung cancer patients, according to Joan H. Schiller, MD.

“This trial illustrates that PD-1 pathway inhibitors can be successfully combined with chemotherapy,” Dr. Schiller wrote in an editorial in the New England Journal of Medicine.

In the study, patients treated with carboplatin-pemetrexed-pembrolizumab therapy had longer overall and progression-free survival compared with patients treated with just chemotherapy.

“The magnitude of benefit is impressive,” Dr. Schiller wrote, “with a hazard ratio for death of 0.49 and 12-month overall survival of 69.2% in the pembrolizumab-combination group.”

However, “many unanswered questions remain,” Dr. Schiller said, particularly concerning the utility of the commonly used PD-L1 tumor proportion score, which “appears to be problematic, with some randomized trials showing benefit only with a score of 50% or more and others showing benefit at other cutoff points, including less than 1%.”

In the current study, all subgroups benefited from the addition of pembrolizumab, including patients with a score of less than 1%.

Despite the need for more research, Dr. Schiller said she believes that existing findings have revealed unprecedented benefits. “Is carboplatin-pemetrexed-pembrolizumab now the first-line standard therapy for patients with nonsquamous NSCLC lacking targetable mutations? In my opinion, the answer is yes.”

Dr. Schiller is with the Inova Schar Cancer Institute. These comments are adapted from an editorial (N Eng J Med. 2018 May 31. doi: 10.1056/NEJMe1804364 ). The author reported financial support from Merck, Lily, AstraZeneca, Genentech/Roche, OncoGenex, Halozyme, Synta, Vertex, Bristol-Myers Squibb, Clovis, Xcovery, AbbVie, Astex, Janssen, and Free to Breathe.

Title
First-line pembrolizumab should be standard
First-line pembrolizumab should be standard

 

In patients with advanced non–small-cell lung cancer (NSCLC) without targetable mutations, the addition of pembrolizumab to first-line combination chemotherapy improves overall and progression-free survival, according to results from the phase 3 KEYNOTE-189 trial.

The researchers found that while patients in all subgroups benefited from the addition of pembrolizumab to chemotherapy, patients with a tumor proportion score of at least 50% benefited the most.

In 2017, accelerated approval was granted for pembrolizumab in combination with carboplatin and pemetrexed as first-line therapy for patients with NSCLC lacking EGFR and ALK mutations. This approval was based on response rates and progression-free survival from the KEYNOTE-024 phase 2 trial.

KEYNOTE-189 solidifies these findings, according to investigators. “Together with the results from KEYNOTE-024, the data from KEYNOTE-189 suggest that introducing immunotherapy as a first-line therapy may have a favorable long-term effect on outcomes,” wrote lead author Leena Gandhi, MD, director of the Perlmutter Cancer Center at New York University, and her colleagues. The report was published in the New England Journal of Medicine.

The double-blind, phase 3 trial assessed 616 treatment-naive patients with metastatic nonsquamous NSCLC without ALK or EGFR mutations. Patients received a platinum-based drug and pemetrexed plus placebo or 200 mg of pembrolizumab every 3 weeks for four cycles. After this, patients received placebo or pembrolizumab with pemetrexed maintenance therapy for up to 35 cycles. Primary endpoints were overall and progression-free survival, determined by a blinded radiologist.

At 12 months, the estimated overall survival was 69.2% for patients treated with pembrolizumab plus chemotherapy, compared with 49.4% for patients treated with chemotherapy alone. For patients treated with the pembrolizumab combination, median progression-free survival was 8.8 months, compared with 4.9 months for those treated with just chemotherapy. Median follow-up time was 10.5 months.

“The survival benefit associated with the pembrolizumab combination was observed in all subgroups of PD-L1 tumor proportion scores,” the authors wrote, “including patients with a score of less than 1%, a population for which single-agent PD-1 and PD-L1 inhibition have a small chance of benefit.” As with previous studies, the subgroup of patients with a tumor proportion score greater than or equal to 50% received the greatest benefit when pembrolizumab was added.


The authors noted that further research is needed to determine whether patients with high PD-L1 expression (TMS ≥ 50%) would benefit more from pembrolizumab combination therapy compared with pembrolizumab monotherapy, which is currently indicated.

SOURCE: Gandhi et al. N Engl J Med. 2018 May 31. doi: 10.1056/NEJMoa1801005.

 

In patients with advanced non–small-cell lung cancer (NSCLC) without targetable mutations, the addition of pembrolizumab to first-line combination chemotherapy improves overall and progression-free survival, according to results from the phase 3 KEYNOTE-189 trial.

The researchers found that while patients in all subgroups benefited from the addition of pembrolizumab to chemotherapy, patients with a tumor proportion score of at least 50% benefited the most.

In 2017, accelerated approval was granted for pembrolizumab in combination with carboplatin and pemetrexed as first-line therapy for patients with NSCLC lacking EGFR and ALK mutations. This approval was based on response rates and progression-free survival from the KEYNOTE-024 phase 2 trial.

KEYNOTE-189 solidifies these findings, according to investigators. “Together with the results from KEYNOTE-024, the data from KEYNOTE-189 suggest that introducing immunotherapy as a first-line therapy may have a favorable long-term effect on outcomes,” wrote lead author Leena Gandhi, MD, director of the Perlmutter Cancer Center at New York University, and her colleagues. The report was published in the New England Journal of Medicine.

The double-blind, phase 3 trial assessed 616 treatment-naive patients with metastatic nonsquamous NSCLC without ALK or EGFR mutations. Patients received a platinum-based drug and pemetrexed plus placebo or 200 mg of pembrolizumab every 3 weeks for four cycles. After this, patients received placebo or pembrolizumab with pemetrexed maintenance therapy for up to 35 cycles. Primary endpoints were overall and progression-free survival, determined by a blinded radiologist.

At 12 months, the estimated overall survival was 69.2% for patients treated with pembrolizumab plus chemotherapy, compared with 49.4% for patients treated with chemotherapy alone. For patients treated with the pembrolizumab combination, median progression-free survival was 8.8 months, compared with 4.9 months for those treated with just chemotherapy. Median follow-up time was 10.5 months.

“The survival benefit associated with the pembrolizumab combination was observed in all subgroups of PD-L1 tumor proportion scores,” the authors wrote, “including patients with a score of less than 1%, a population for which single-agent PD-1 and PD-L1 inhibition have a small chance of benefit.” As with previous studies, the subgroup of patients with a tumor proportion score greater than or equal to 50% received the greatest benefit when pembrolizumab was added.


The authors noted that further research is needed to determine whether patients with high PD-L1 expression (TMS ≥ 50%) would benefit more from pembrolizumab combination therapy compared with pembrolizumab monotherapy, which is currently indicated.

SOURCE: Gandhi et al. N Engl J Med. 2018 May 31. doi: 10.1056/NEJMoa1801005.

Publications
Publications
Topics
Article Type
Click for Credit Status
Ready
Sections
Article Source

FROM THE NEW ENGLAND JOURNAL OF MEDICINE

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Vitals

 

Key clinical point: In patients with advanced NSCLC without targetable mutations, the addition of pembrolizumab to standard combination chemotherapy improves overall survival and progression-free survival.

Major finding: The estimated rate of overall survival at 12 months in patients treated with pembrolizumab-combination therapy was 69.2% (95% CI, 64.1-73.8) compared with 49.4% (95% CI, 42.1-56.2) in patients treated with placebo-combination therapy.

Study details: A double-blind, phase 3 trial of 616 patients with metastatic NSCLC lacking ALK or EGFR mutations (KEYNOTE-189).

Disclosures: Merck sponsored the study. Researchers reported financial support from Genentech/Roche, Pfizer, Ignyta, AbbVie, Eli Lilly, and other companies.

Source: Gandhi et al. N Engl J Med. 2018 May 31. doi: 10.1056/NEJMoa1801005.

Disqus Comments
Default
Use ProPublica

Compared With Interferon, Fingolimod Improves MRI Outcomes of Pediatric MS

Article Type
Changed
Wed, 01/16/2019 - 15:38
Fingolimod slows brain volume loss for two years, compared with interferon beta-1a.

NASHVILLE—In patients with pediatric-onset multiple sclerosis (MS), fingolimod significantly reduces MRI activity and slows brain volume loss for as long as two years, compared with interferon beta-1a, according to data described at the 2018 CMSC Annual Meeting.

Tanuja Chitnis, MD
Between 3% and 5% of cases of MS manifest in childhood and adolescence, characteristically with a highly active inflammatory disease course. Pediatric-onset MS reduces brain integrity by disrupting tissue microstructure. The disease also may increase brain volume loss above age-expected rates. Tanuja Chitnis, MD, Director of the Partners Pediatric MS Center at Massachusetts General Hospital in Boston, and colleagues analyzed data from the PARADIGMS trial to assess the effect of fingolimod (in doses as high as 0.5 mg/day) versus intramuscular interferon beta-1a (30 μg once weekly) on predefined MRI outcomes in patients with pediatric-onset MS. 

Analyzing the PARADIGMS Data

PARADIGMS was a double-blind, double-dummy, active-controlled, parallel-group, multicenter study in which patients participated for as long as two years. The investigators randomized patients with pediatric-onset MS (ages 10 through 17) to oral fingolimod or interferon beta-1a. The dose of fingolimod was adjusted for body weight. MRI was performed at baseline and every six months thereafter until the end of the study core phase. A central reading center analyzed the MRI results. The key MRI outcomes were the number of new or newly enlarged T2 lesions and gadolinium-enhancing T1 lesions, annual rate of brain volume change, number of new T1 hypointense lesions, change in total T2 hyperintense lesion volume, and number of combined unique active lesions.

The researchers randomized 107 participants to oral fingolimod and 108 to interferon beta-1a. At baseline, mean age was about 15. Most patients were female. Mean disease duration was one to two years. The average number of relapses in the year before screening was approximately 1.5. Participants had 2.6 to 3.1 gadolinium-enhancing T1 lesions.

Data Support Fingolimod’s Efficacy in Pediatric MS

At the end of the study, fingolimod significantly reduced the annualized rate of new or newly enlarged T2 lesions by 52.6% and the number of gadolinium-enhancing T1 lesions per scan by 66.0%, compared with interferon beta-1a. The odds ratio of freedom from new or newly enlarged T2 lesions was 4.51 in the fingolimod arm, compared with the interferon beta-1a arm. The odds ratio of freedom from gadolinium-enhancing lesions was 3.0 in the fingolimod arm, compared with the interferon beta-1a arm.

Compared with interferon beta-1a, treatment with fingolimod for as long as two years significantly reduced the annualized rate of brain volume change (least squares mean: 0.48 vs 0.80). Fingolimod reduced the annualized rate of new T1 hypointense lesions by 62.8% and the number of combined unique active lesions per scan by 60.7%, compared with interferon beta-1a. Fingolimod also reduced T2 hyperintense lesion volume, compared with interferon beta-1a (percentage change from baseline: 18.4% vs 32.4%).

“The rate of T2-related atrophy [in pediatric MS] is concerning, and I am particularly interested in looking at the extension study data as they come out, and we’ll see if there is a decrease during longer-term treatment,” said Dr. Chitnis.

“These results, overall, along with the efficacy demonstrated on the clinical relapse rate, support the overall benefit of fingolimod in pediatric patients with MS,” she concluded.

—Erik Greb

Publications
Topics
Sections
Fingolimod slows brain volume loss for two years, compared with interferon beta-1a.
Fingolimod slows brain volume loss for two years, compared with interferon beta-1a.

NASHVILLE—In patients with pediatric-onset multiple sclerosis (MS), fingolimod significantly reduces MRI activity and slows brain volume loss for as long as two years, compared with interferon beta-1a, according to data described at the 2018 CMSC Annual Meeting.

Tanuja Chitnis, MD
Between 3% and 5% of cases of MS manifest in childhood and adolescence, characteristically with a highly active inflammatory disease course. Pediatric-onset MS reduces brain integrity by disrupting tissue microstructure. The disease also may increase brain volume loss above age-expected rates. Tanuja Chitnis, MD, Director of the Partners Pediatric MS Center at Massachusetts General Hospital in Boston, and colleagues analyzed data from the PARADIGMS trial to assess the effect of fingolimod (in doses as high as 0.5 mg/day) versus intramuscular interferon beta-1a (30 μg once weekly) on predefined MRI outcomes in patients with pediatric-onset MS. 

Analyzing the PARADIGMS Data

PARADIGMS was a double-blind, double-dummy, active-controlled, parallel-group, multicenter study in which patients participated for as long as two years. The investigators randomized patients with pediatric-onset MS (ages 10 through 17) to oral fingolimod or interferon beta-1a. The dose of fingolimod was adjusted for body weight. MRI was performed at baseline and every six months thereafter until the end of the study core phase. A central reading center analyzed the MRI results. The key MRI outcomes were the number of new or newly enlarged T2 lesions and gadolinium-enhancing T1 lesions, annual rate of brain volume change, number of new T1 hypointense lesions, change in total T2 hyperintense lesion volume, and number of combined unique active lesions.

The researchers randomized 107 participants to oral fingolimod and 108 to interferon beta-1a. At baseline, mean age was about 15. Most patients were female. Mean disease duration was one to two years. The average number of relapses in the year before screening was approximately 1.5. Participants had 2.6 to 3.1 gadolinium-enhancing T1 lesions.

Data Support Fingolimod’s Efficacy in Pediatric MS

At the end of the study, fingolimod significantly reduced the annualized rate of new or newly enlarged T2 lesions by 52.6% and the number of gadolinium-enhancing T1 lesions per scan by 66.0%, compared with interferon beta-1a. The odds ratio of freedom from new or newly enlarged T2 lesions was 4.51 in the fingolimod arm, compared with the interferon beta-1a arm. The odds ratio of freedom from gadolinium-enhancing lesions was 3.0 in the fingolimod arm, compared with the interferon beta-1a arm.

Compared with interferon beta-1a, treatment with fingolimod for as long as two years significantly reduced the annualized rate of brain volume change (least squares mean: 0.48 vs 0.80). Fingolimod reduced the annualized rate of new T1 hypointense lesions by 62.8% and the number of combined unique active lesions per scan by 60.7%, compared with interferon beta-1a. Fingolimod also reduced T2 hyperintense lesion volume, compared with interferon beta-1a (percentage change from baseline: 18.4% vs 32.4%).

“The rate of T2-related atrophy [in pediatric MS] is concerning, and I am particularly interested in looking at the extension study data as they come out, and we’ll see if there is a decrease during longer-term treatment,” said Dr. Chitnis.

“These results, overall, along with the efficacy demonstrated on the clinical relapse rate, support the overall benefit of fingolimod in pediatric patients with MS,” she concluded.

—Erik Greb

NASHVILLE—In patients with pediatric-onset multiple sclerosis (MS), fingolimod significantly reduces MRI activity and slows brain volume loss for as long as two years, compared with interferon beta-1a, according to data described at the 2018 CMSC Annual Meeting.

Tanuja Chitnis, MD
Between 3% and 5% of cases of MS manifest in childhood and adolescence, characteristically with a highly active inflammatory disease course. Pediatric-onset MS reduces brain integrity by disrupting tissue microstructure. The disease also may increase brain volume loss above age-expected rates. Tanuja Chitnis, MD, Director of the Partners Pediatric MS Center at Massachusetts General Hospital in Boston, and colleagues analyzed data from the PARADIGMS trial to assess the effect of fingolimod (in doses as high as 0.5 mg/day) versus intramuscular interferon beta-1a (30 μg once weekly) on predefined MRI outcomes in patients with pediatric-onset MS. 

Analyzing the PARADIGMS Data

PARADIGMS was a double-blind, double-dummy, active-controlled, parallel-group, multicenter study in which patients participated for as long as two years. The investigators randomized patients with pediatric-onset MS (ages 10 through 17) to oral fingolimod or interferon beta-1a. The dose of fingolimod was adjusted for body weight. MRI was performed at baseline and every six months thereafter until the end of the study core phase. A central reading center analyzed the MRI results. The key MRI outcomes were the number of new or newly enlarged T2 lesions and gadolinium-enhancing T1 lesions, annual rate of brain volume change, number of new T1 hypointense lesions, change in total T2 hyperintense lesion volume, and number of combined unique active lesions.

The researchers randomized 107 participants to oral fingolimod and 108 to interferon beta-1a. At baseline, mean age was about 15. Most patients were female. Mean disease duration was one to two years. The average number of relapses in the year before screening was approximately 1.5. Participants had 2.6 to 3.1 gadolinium-enhancing T1 lesions.

Data Support Fingolimod’s Efficacy in Pediatric MS

At the end of the study, fingolimod significantly reduced the annualized rate of new or newly enlarged T2 lesions by 52.6% and the number of gadolinium-enhancing T1 lesions per scan by 66.0%, compared with interferon beta-1a. The odds ratio of freedom from new or newly enlarged T2 lesions was 4.51 in the fingolimod arm, compared with the interferon beta-1a arm. The odds ratio of freedom from gadolinium-enhancing lesions was 3.0 in the fingolimod arm, compared with the interferon beta-1a arm.

Compared with interferon beta-1a, treatment with fingolimod for as long as two years significantly reduced the annualized rate of brain volume change (least squares mean: 0.48 vs 0.80). Fingolimod reduced the annualized rate of new T1 hypointense lesions by 62.8% and the number of combined unique active lesions per scan by 60.7%, compared with interferon beta-1a. Fingolimod also reduced T2 hyperintense lesion volume, compared with interferon beta-1a (percentage change from baseline: 18.4% vs 32.4%).

“The rate of T2-related atrophy [in pediatric MS] is concerning, and I am particularly interested in looking at the extension study data as they come out, and we’ll see if there is a decrease during longer-term treatment,” said Dr. Chitnis.

“These results, overall, along with the efficacy demonstrated on the clinical relapse rate, support the overall benefit of fingolimod in pediatric patients with MS,” she concluded.

—Erik Greb

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica

Can Exercise Improve Vision in Children With MS?

Article Type
Changed
Wed, 01/16/2019 - 15:38

A positive association was observed between moderate-to-vigorous physical activity and retinal nerve fiber layer thickness in pediatric patients.

NASHVILLE—Moderate-to-vigorous physical activity is positively associated with retinal nerve fiber layer thickness in children with multiple sclerosis (MS), according to research presented at the 2018 CMSC Annual Meeting. This finding may help to support an intervention targeting moderate-to-vigorous physical activity to improve anterior visual pathway integrity in children with MS.

More than one-third of pediatric patients with MS experience optic neuritis, and most experience visual pathway abnormalities, including reductions in the retinal nerve fiber layer and ganglion cell inner-plexiform layer. Previous studies in adults have shown a positive association between moderate-to-vigorous physical activity and the retinal nerve fiber layer; however, this association has not been evaluated in pediatric patients with MS.

To investigate the associations between mild-to-vigorous physical activity, the retinal nerve fiber layer, and the ganglion cell inner-plexiform layer in pediatric patients with MS, Alexander L. Pearson, a medical student at the University of Ottawa in Ontario, and colleagues conducted a cross-sectional study.

The researchers recruited participants from the Pediatric MS and Demyelinating Disorders Center at the Hospital for Sick Children in Toronto. Eligible participants had a diagnosis of MS (according to the International Pediatric MS Study Group consensus definitions) and were younger than 18. Patients with neuroinflammatory abnormalities associated with underlying systemic or neurologic disorders, recurrent neuroinflammatory disorders other than MS, coexisting ocular pathologies, visual acuity ±6 diopters or worse, were excluded.

Participants received standardized visual evaluations, including ocular coherence tomography. Investigators performed evaluations more than 90 days after an optic neuritis episode using a spectral-domain ocular coherence tomography Cirrus scanner. Participants also completed the Godin Leisure-Time Exercise Questionnaire (GLTEQ) more than 30 days after a relapse. This questionnaire was used to calculate the health contribution score.

Generalized linear models were used to assess the associations between moderate-to-vigorous physical activity, the retinal nerve fiber layer, and the ganglion cell inner-plexiform layer when controlling for sex, number of optic neuritis episodes, disease duration at time of ocular coherence tomography, and within-subject correlation between eyes. Bonferroni correction was used to adjust for multiple comparisons.

Thirty patients participated in this study; 23 were female. Ocular coherence tomography was performed at a mean age of 15.7 (range, 10.6–18.0) and a median of 1.9 years from disease onset. The median retinal nerve fiber layer was 90 μm, and the median ganglion cell inner-plexiform layer was 73.5 μm. The median amount of moderate-to-vigorous physical activity was 26.5 metabolic equivalents per week.

The research team found that moderate-to-vigorous physical activity was positively associated with retinal nerve fiber layer thickness. Although the retinal nerve fiber layer and ganglion cell inner-plexiform layer were moderately correlated, moderate-to-vigorous physical activity was not associated with the ganglion cell inner-plexiform layer, said the authors.

“Next steps include a trial using mild-to-vigorous physical activity to improve anterior visual pathway integrity in children with MS,” the researchers concluded.

Issue
Neurology Reviews - 26(7)
Publications
Topics
Page Number
32
Sections

A positive association was observed between moderate-to-vigorous physical activity and retinal nerve fiber layer thickness in pediatric patients.

A positive association was observed between moderate-to-vigorous physical activity and retinal nerve fiber layer thickness in pediatric patients.

NASHVILLE—Moderate-to-vigorous physical activity is positively associated with retinal nerve fiber layer thickness in children with multiple sclerosis (MS), according to research presented at the 2018 CMSC Annual Meeting. This finding may help to support an intervention targeting moderate-to-vigorous physical activity to improve anterior visual pathway integrity in children with MS.

More than one-third of pediatric patients with MS experience optic neuritis, and most experience visual pathway abnormalities, including reductions in the retinal nerve fiber layer and ganglion cell inner-plexiform layer. Previous studies in adults have shown a positive association between moderate-to-vigorous physical activity and the retinal nerve fiber layer; however, this association has not been evaluated in pediatric patients with MS.

To investigate the associations between mild-to-vigorous physical activity, the retinal nerve fiber layer, and the ganglion cell inner-plexiform layer in pediatric patients with MS, Alexander L. Pearson, a medical student at the University of Ottawa in Ontario, and colleagues conducted a cross-sectional study.

The researchers recruited participants from the Pediatric MS and Demyelinating Disorders Center at the Hospital for Sick Children in Toronto. Eligible participants had a diagnosis of MS (according to the International Pediatric MS Study Group consensus definitions) and were younger than 18. Patients with neuroinflammatory abnormalities associated with underlying systemic or neurologic disorders, recurrent neuroinflammatory disorders other than MS, coexisting ocular pathologies, visual acuity ±6 diopters or worse, were excluded.

Participants received standardized visual evaluations, including ocular coherence tomography. Investigators performed evaluations more than 90 days after an optic neuritis episode using a spectral-domain ocular coherence tomography Cirrus scanner. Participants also completed the Godin Leisure-Time Exercise Questionnaire (GLTEQ) more than 30 days after a relapse. This questionnaire was used to calculate the health contribution score.

Generalized linear models were used to assess the associations between moderate-to-vigorous physical activity, the retinal nerve fiber layer, and the ganglion cell inner-plexiform layer when controlling for sex, number of optic neuritis episodes, disease duration at time of ocular coherence tomography, and within-subject correlation between eyes. Bonferroni correction was used to adjust for multiple comparisons.

Thirty patients participated in this study; 23 were female. Ocular coherence tomography was performed at a mean age of 15.7 (range, 10.6–18.0) and a median of 1.9 years from disease onset. The median retinal nerve fiber layer was 90 μm, and the median ganglion cell inner-plexiform layer was 73.5 μm. The median amount of moderate-to-vigorous physical activity was 26.5 metabolic equivalents per week.

The research team found that moderate-to-vigorous physical activity was positively associated with retinal nerve fiber layer thickness. Although the retinal nerve fiber layer and ganglion cell inner-plexiform layer were moderately correlated, moderate-to-vigorous physical activity was not associated with the ganglion cell inner-plexiform layer, said the authors.

“Next steps include a trial using mild-to-vigorous physical activity to improve anterior visual pathway integrity in children with MS,” the researchers concluded.

NASHVILLE—Moderate-to-vigorous physical activity is positively associated with retinal nerve fiber layer thickness in children with multiple sclerosis (MS), according to research presented at the 2018 CMSC Annual Meeting. This finding may help to support an intervention targeting moderate-to-vigorous physical activity to improve anterior visual pathway integrity in children with MS.

More than one-third of pediatric patients with MS experience optic neuritis, and most experience visual pathway abnormalities, including reductions in the retinal nerve fiber layer and ganglion cell inner-plexiform layer. Previous studies in adults have shown a positive association between moderate-to-vigorous physical activity and the retinal nerve fiber layer; however, this association has not been evaluated in pediatric patients with MS.

To investigate the associations between mild-to-vigorous physical activity, the retinal nerve fiber layer, and the ganglion cell inner-plexiform layer in pediatric patients with MS, Alexander L. Pearson, a medical student at the University of Ottawa in Ontario, and colleagues conducted a cross-sectional study.

The researchers recruited participants from the Pediatric MS and Demyelinating Disorders Center at the Hospital for Sick Children in Toronto. Eligible participants had a diagnosis of MS (according to the International Pediatric MS Study Group consensus definitions) and were younger than 18. Patients with neuroinflammatory abnormalities associated with underlying systemic or neurologic disorders, recurrent neuroinflammatory disorders other than MS, coexisting ocular pathologies, visual acuity ±6 diopters or worse, were excluded.

Participants received standardized visual evaluations, including ocular coherence tomography. Investigators performed evaluations more than 90 days after an optic neuritis episode using a spectral-domain ocular coherence tomography Cirrus scanner. Participants also completed the Godin Leisure-Time Exercise Questionnaire (GLTEQ) more than 30 days after a relapse. This questionnaire was used to calculate the health contribution score.

Generalized linear models were used to assess the associations between moderate-to-vigorous physical activity, the retinal nerve fiber layer, and the ganglion cell inner-plexiform layer when controlling for sex, number of optic neuritis episodes, disease duration at time of ocular coherence tomography, and within-subject correlation between eyes. Bonferroni correction was used to adjust for multiple comparisons.

Thirty patients participated in this study; 23 were female. Ocular coherence tomography was performed at a mean age of 15.7 (range, 10.6–18.0) and a median of 1.9 years from disease onset. The median retinal nerve fiber layer was 90 μm, and the median ganglion cell inner-plexiform layer was 73.5 μm. The median amount of moderate-to-vigorous physical activity was 26.5 metabolic equivalents per week.

The research team found that moderate-to-vigorous physical activity was positively associated with retinal nerve fiber layer thickness. Although the retinal nerve fiber layer and ganglion cell inner-plexiform layer were moderately correlated, moderate-to-vigorous physical activity was not associated with the ganglion cell inner-plexiform layer, said the authors.

“Next steps include a trial using mild-to-vigorous physical activity to improve anterior visual pathway integrity in children with MS,” the researchers concluded.

Issue
Neurology Reviews - 26(7)
Issue
Neurology Reviews - 26(7)
Page Number
32
Page Number
32
Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica

More Frequent Dosing of Interferon Beta-1a May Benefit Patients With MS With Breakthrough Disease

Article Type
Changed
Wed, 01/16/2019 - 15:38
Twenty-six patients receiving twice-weekly treatment had no further breakthrough disease for 14 months or more.

NASHVILLE—Patients with multiple sclerosis (MS) with breakthrough disease may benefit from intramuscular interferon beta 1-a treatment twice per week, according to research described at the 2018 CMSC Annual Meeting. “Advantages to using an intramuscular interferon beta 1-a preparation include no skin reactions and a lower incidence of interferon neutralizing antibodies,” said Robert W. Baumhefner, MD, a neurologist at the Veteran Affairs West Los Angeles Medical Center.

Previous clinical trials have suggested a dose-response effect for interferon beta in MS. The European Interferon Beta-1a Dose Comparison Study, however, found no change in efficacy with just doubling the standard dose of intramuscular interferon beta-1a once per week. This may not be the same as increasing the frequency of intramuscular interferon administration, said Baumhefner. In addition, none of the previous studies have information on patients with breakthrough disease on standard-dose intramuscular interferon beta-1a switched to twice-weekly dosing.

Dr. Baumhefner conducted a retrospective observational study of patients MS with breakthrough disease receiving intramuscular interferon beta 1-a once per week who were switched to intramuscular interferon beta 1-a twice per week.

A total of 107 patients with MS were started on intramuscular interferon beta 1-a from 1995 to 2015 at the MS clinic of the VA West Los Angeles Medical Center. Of these, 59 patients with breakthrough disease were switched to twice-weekly intramuscular interferon beta-1a. There was adequate follow-up for at least two years for 52 of these patients. In addition, participants were followed up an average of every four months.

At each visit, an interval history of any relapse; scores on the Incapacity Status Scale, Functional Systems Scale, and Expanded Disability Status Scale (EDSS); and a proprietary graded neurologic examination were obtained. Annual MRI of the brain using a contrast-enhanced MS protocol was obtained in most patients. Baumhefner defined breakthrough disease as continued clinical relapses, new T2 or enhanced lesions on MRI, or worsening of EDSS or neurologic examination.

Of the 52 patients with adequate follow-up, 26 had no further breakthrough disease for 14 months or longer (range, 14-192 months). Five patients did not tolerate the increase in frequency of administration. Interferon beta neutralizing antibody testing was performed on 25 patients while they were receiving twice-weekly dosing. One patient who failed twice-weekly interferon beta had consistently elevated titers on two determinations (4%). African American patients, those with a higher EDSS score when switching, and patients with a longer duration of stability on weekly dosed treatment may be less likely to respond, the researcher concluded.

Publications
Topics
Sections
Twenty-six patients receiving twice-weekly treatment had no further breakthrough disease for 14 months or more.
Twenty-six patients receiving twice-weekly treatment had no further breakthrough disease for 14 months or more.

NASHVILLE—Patients with multiple sclerosis (MS) with breakthrough disease may benefit from intramuscular interferon beta 1-a treatment twice per week, according to research described at the 2018 CMSC Annual Meeting. “Advantages to using an intramuscular interferon beta 1-a preparation include no skin reactions and a lower incidence of interferon neutralizing antibodies,” said Robert W. Baumhefner, MD, a neurologist at the Veteran Affairs West Los Angeles Medical Center.

Previous clinical trials have suggested a dose-response effect for interferon beta in MS. The European Interferon Beta-1a Dose Comparison Study, however, found no change in efficacy with just doubling the standard dose of intramuscular interferon beta-1a once per week. This may not be the same as increasing the frequency of intramuscular interferon administration, said Baumhefner. In addition, none of the previous studies have information on patients with breakthrough disease on standard-dose intramuscular interferon beta-1a switched to twice-weekly dosing.

Dr. Baumhefner conducted a retrospective observational study of patients MS with breakthrough disease receiving intramuscular interferon beta 1-a once per week who were switched to intramuscular interferon beta 1-a twice per week.

A total of 107 patients with MS were started on intramuscular interferon beta 1-a from 1995 to 2015 at the MS clinic of the VA West Los Angeles Medical Center. Of these, 59 patients with breakthrough disease were switched to twice-weekly intramuscular interferon beta-1a. There was adequate follow-up for at least two years for 52 of these patients. In addition, participants were followed up an average of every four months.

At each visit, an interval history of any relapse; scores on the Incapacity Status Scale, Functional Systems Scale, and Expanded Disability Status Scale (EDSS); and a proprietary graded neurologic examination were obtained. Annual MRI of the brain using a contrast-enhanced MS protocol was obtained in most patients. Baumhefner defined breakthrough disease as continued clinical relapses, new T2 or enhanced lesions on MRI, or worsening of EDSS or neurologic examination.

Of the 52 patients with adequate follow-up, 26 had no further breakthrough disease for 14 months or longer (range, 14-192 months). Five patients did not tolerate the increase in frequency of administration. Interferon beta neutralizing antibody testing was performed on 25 patients while they were receiving twice-weekly dosing. One patient who failed twice-weekly interferon beta had consistently elevated titers on two determinations (4%). African American patients, those with a higher EDSS score when switching, and patients with a longer duration of stability on weekly dosed treatment may be less likely to respond, the researcher concluded.

NASHVILLE—Patients with multiple sclerosis (MS) with breakthrough disease may benefit from intramuscular interferon beta 1-a treatment twice per week, according to research described at the 2018 CMSC Annual Meeting. “Advantages to using an intramuscular interferon beta 1-a preparation include no skin reactions and a lower incidence of interferon neutralizing antibodies,” said Robert W. Baumhefner, MD, a neurologist at the Veteran Affairs West Los Angeles Medical Center.

Previous clinical trials have suggested a dose-response effect for interferon beta in MS. The European Interferon Beta-1a Dose Comparison Study, however, found no change in efficacy with just doubling the standard dose of intramuscular interferon beta-1a once per week. This may not be the same as increasing the frequency of intramuscular interferon administration, said Baumhefner. In addition, none of the previous studies have information on patients with breakthrough disease on standard-dose intramuscular interferon beta-1a switched to twice-weekly dosing.

Dr. Baumhefner conducted a retrospective observational study of patients MS with breakthrough disease receiving intramuscular interferon beta 1-a once per week who were switched to intramuscular interferon beta 1-a twice per week.

A total of 107 patients with MS were started on intramuscular interferon beta 1-a from 1995 to 2015 at the MS clinic of the VA West Los Angeles Medical Center. Of these, 59 patients with breakthrough disease were switched to twice-weekly intramuscular interferon beta-1a. There was adequate follow-up for at least two years for 52 of these patients. In addition, participants were followed up an average of every four months.

At each visit, an interval history of any relapse; scores on the Incapacity Status Scale, Functional Systems Scale, and Expanded Disability Status Scale (EDSS); and a proprietary graded neurologic examination were obtained. Annual MRI of the brain using a contrast-enhanced MS protocol was obtained in most patients. Baumhefner defined breakthrough disease as continued clinical relapses, new T2 or enhanced lesions on MRI, or worsening of EDSS or neurologic examination.

Of the 52 patients with adequate follow-up, 26 had no further breakthrough disease for 14 months or longer (range, 14-192 months). Five patients did not tolerate the increase in frequency of administration. Interferon beta neutralizing antibody testing was performed on 25 patients while they were receiving twice-weekly dosing. One patient who failed twice-weekly interferon beta had consistently elevated titers on two determinations (4%). African American patients, those with a higher EDSS score when switching, and patients with a longer duration of stability on weekly dosed treatment may be less likely to respond, the researcher concluded.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica