User login
Oral contraceptive efficacy unaffected by dimethyl fumarate
NATIONAL HARBOR, MD. – Delayed-release dimethyl fumarate (DMF) coadministered with norgestimate/ethinyl estradiol, a commonly used progesterone-estrogen combination oral contraceptive, did not alter the OC’s pharmacokinetics or pharmacodynamics, according to the results of a small study.
The safety profile of the coadministered preparation is similar to the profile of DMF (Tecfidera, Biogen) alone, Dr. Bing Zhu said in a poster session at the annual meeting of the Consortium of Multiple Sclerosis Centers.
The findings indicate that women with relapsing-remitting multiple sclerosis (RRMS) who are of childbearing age and who are being treated with DMF can use OCs without having to modify the contraceptive dose, said Dr. Zhu, who is an employee of Biogen, Cambridge, Mass.
The study involved healthy women aged 18-45 years (mean age about 31 years) who were able to conceive. All received a daily OC (Ortho-Cyclen; 250 mcg norgestimate, 35 mcg ethinyl estradiol). After 28 days, those with progesterone levels less than 3 ng/mL were randomized to a 28-day regimen of either the daily dose of OC (n = 39) or OC along with DMF (240 mg twice daily), designated period 1, with crossover to the other treatment for a further 28 days (period 2). Blood samples were collected during the first 24 hours for pharmacokinetic measurements and at 2, 3, and 4 weeks for pharmacodynamic determinations.
The primary objective was the pharmacokinetics of norgestimate, as determined by measuring the levels of its main metabolite, norelgestromin. Secondary objectives included pharmacodynamics, as determined by the levels of serum progesterone and the safety/tolerability of DMF.
Plasma concentrations of norelgestromin and ethinyl estradiol were identical over time in the OC and OC + DMF groups in period 1, as was serum progesterone.
Treatment-emergent adverse events were similar in type and severity in both groups. Most were mild. The prevalent adverse events included flushing and gastrointestinal disorders accompanied by nausea and vomiting. Adverse events occurred in 10 of the 39 (26%) subjects who received the OC and in 26 of the 39 (67%) subjects who received the OC + DMF. Discontinuation because of adverse events occurred in 8% and 15% of subjects who received the OC and the OC + DMF, respectively. There were no deaths.
“The results suggest that women of childbearing potential treated with DMF are able to use a combined OC for contraception without dose modification,” said Dr. Zhu.
The study findings were recently published (Neurology. 2016 Apr 5;86:Suppl P2.097).
The study was funded by Biogen. Dr. Zhu is a Biogen employee.
NATIONAL HARBOR, MD. – Delayed-release dimethyl fumarate (DMF) coadministered with norgestimate/ethinyl estradiol, a commonly used progesterone-estrogen combination oral contraceptive, did not alter the OC’s pharmacokinetics or pharmacodynamics, according to the results of a small study.
The safety profile of the coadministered preparation is similar to the profile of DMF (Tecfidera, Biogen) alone, Dr. Bing Zhu said in a poster session at the annual meeting of the Consortium of Multiple Sclerosis Centers.
The findings indicate that women with relapsing-remitting multiple sclerosis (RRMS) who are of childbearing age and who are being treated with DMF can use OCs without having to modify the contraceptive dose, said Dr. Zhu, who is an employee of Biogen, Cambridge, Mass.
The study involved healthy women aged 18-45 years (mean age about 31 years) who were able to conceive. All received a daily OC (Ortho-Cyclen; 250 mcg norgestimate, 35 mcg ethinyl estradiol). After 28 days, those with progesterone levels less than 3 ng/mL were randomized to a 28-day regimen of either the daily dose of OC (n = 39) or OC along with DMF (240 mg twice daily), designated period 1, with crossover to the other treatment for a further 28 days (period 2). Blood samples were collected during the first 24 hours for pharmacokinetic measurements and at 2, 3, and 4 weeks for pharmacodynamic determinations.
The primary objective was the pharmacokinetics of norgestimate, as determined by measuring the levels of its main metabolite, norelgestromin. Secondary objectives included pharmacodynamics, as determined by the levels of serum progesterone and the safety/tolerability of DMF.
Plasma concentrations of norelgestromin and ethinyl estradiol were identical over time in the OC and OC + DMF groups in period 1, as was serum progesterone.
Treatment-emergent adverse events were similar in type and severity in both groups. Most were mild. The prevalent adverse events included flushing and gastrointestinal disorders accompanied by nausea and vomiting. Adverse events occurred in 10 of the 39 (26%) subjects who received the OC and in 26 of the 39 (67%) subjects who received the OC + DMF. Discontinuation because of adverse events occurred in 8% and 15% of subjects who received the OC and the OC + DMF, respectively. There were no deaths.
“The results suggest that women of childbearing potential treated with DMF are able to use a combined OC for contraception without dose modification,” said Dr. Zhu.
The study findings were recently published (Neurology. 2016 Apr 5;86:Suppl P2.097).
The study was funded by Biogen. Dr. Zhu is a Biogen employee.
NATIONAL HARBOR, MD. – Delayed-release dimethyl fumarate (DMF) coadministered with norgestimate/ethinyl estradiol, a commonly used progesterone-estrogen combination oral contraceptive, did not alter the OC’s pharmacokinetics or pharmacodynamics, according to the results of a small study.
The safety profile of the coadministered preparation is similar to the profile of DMF (Tecfidera, Biogen) alone, Dr. Bing Zhu said in a poster session at the annual meeting of the Consortium of Multiple Sclerosis Centers.
The findings indicate that women with relapsing-remitting multiple sclerosis (RRMS) who are of childbearing age and who are being treated with DMF can use OCs without having to modify the contraceptive dose, said Dr. Zhu, who is an employee of Biogen, Cambridge, Mass.
The study involved healthy women aged 18-45 years (mean age about 31 years) who were able to conceive. All received a daily OC (Ortho-Cyclen; 250 mcg norgestimate, 35 mcg ethinyl estradiol). After 28 days, those with progesterone levels less than 3 ng/mL were randomized to a 28-day regimen of either the daily dose of OC (n = 39) or OC along with DMF (240 mg twice daily), designated period 1, with crossover to the other treatment for a further 28 days (period 2). Blood samples were collected during the first 24 hours for pharmacokinetic measurements and at 2, 3, and 4 weeks for pharmacodynamic determinations.
The primary objective was the pharmacokinetics of norgestimate, as determined by measuring the levels of its main metabolite, norelgestromin. Secondary objectives included pharmacodynamics, as determined by the levels of serum progesterone and the safety/tolerability of DMF.
Plasma concentrations of norelgestromin and ethinyl estradiol were identical over time in the OC and OC + DMF groups in period 1, as was serum progesterone.
Treatment-emergent adverse events were similar in type and severity in both groups. Most were mild. The prevalent adverse events included flushing and gastrointestinal disorders accompanied by nausea and vomiting. Adverse events occurred in 10 of the 39 (26%) subjects who received the OC and in 26 of the 39 (67%) subjects who received the OC + DMF. Discontinuation because of adverse events occurred in 8% and 15% of subjects who received the OC and the OC + DMF, respectively. There were no deaths.
“The results suggest that women of childbearing potential treated with DMF are able to use a combined OC for contraception without dose modification,” said Dr. Zhu.
The study findings were recently published (Neurology. 2016 Apr 5;86:Suppl P2.097).
The study was funded by Biogen. Dr. Zhu is a Biogen employee.
AT THE CMSC ANNUAL MEETING
Key clinical point: Delayed-release dimethyl fumarate can be used in childbearing women with RRMS without fear of compromising the effect of a common oral contraceptive.
Major finding: DMF coadministered with a common oral contraceptive does not affect the pharmacokinetics and pharmacodynamics of the contraceptive.
Data source: Open-label, randomized, two-way crossover study involving 46 healthy women.
Disclosures: The study was funded by Biogen. Dr. Zhu is an employee of and stockholder in Biogen.
VIDEO: Smoking, excess weight hinder sustained remission in early RA
LONDON – Tobacco use and excess weight can make it harder to achieve sustained remission in the treatment of early rheumatoid arthritis, according to findings from more than 1,000 patients in the Canadian Early Arthritis Cohort (CATCH) multicenter, prospective study.
Aggressive treatment that starts soon after diagnosis of rheumatoid arthritis (RA) is important for the absence of disease activity, which is the hallmark of sustained remission. But the reality is a success rate of less than 50% in the first 3 years with physical deterioration continuing thereafter. “Excess weight and smoking are two risk factors for developing RA. We were interested in seeing if they might also affect how well people responded to treatment,” said Susan Bartlett, Ph.D., a clinical psychologist at McGill University in Montreal.
At the annual European Congress of Rheumatology, Dr. Bartlett and colleagues reported on a cohort of 1,008 early RA patients who were enrolled in the Canadian Early Arthritis Cohort (CATCH) multicenter, prospective study and followed from around the time of diagnosis through the first 3 years of treatment to estimate the time it took until they achieved sustained remission, defined as having a 28-joint Disease Activity Score less than 2.6 for two consecutive visits.
Mean age of the patients (72% female, 81% white) was early 50s. Overall, 30% of females and 47% of males were overweight, one-third of both genders were obese, and 15%-20% smoked. Treatment at entry included methotrexate in mono- or combination therapy in about three-quarters of the patients, with steroids used in about half and biologics used sparingly.
The proportion of patients in sustained remission was 38% at 3 years, with a median time to remission of 11.3 months. “That finding wasn’t surprising because that is generally what is found in most studies of early RA. However, when we looked more closely at who was and wasn’t achieving remission, we found that people who smoked and those who were overweight or obese were much less likely than their nonsmoking, normal-weight peers to be in sustained remission,” Dr. Bartlett said in a pre-congress interview.
After adjustment for factors that could affect response to treatment – including age, race, disability status, pain, and early medications used – smoking (P = .046) and excess weight (P = .003) were associated with a poorer likelihood of achieving sustained remission. While more men than women were overweight or obese, the effects of weight and smoking appeared to be more problematic for women (P = .02).
An average nonsmoking male with a healthy body mass index (BMI; 25 kg/m2 or less) had about a 41% probability of achieving sustained remission within 3 years, compared with 15% for an obese male smoker. A nonsmoking female with a healthy BMI had a 27% probability of achieving sustained remission within 3 years, compared with 10% for an obese female smoker. Probabilities of sustained remission were also lower for overweight men and women, Dr. Bartlett reported.
Smoking and obesity have already been linked with an increased likelihood of developing RA, which in turn increases the risk of cardiovascular disease and premature death. The latest data suggest that both smoking and extra weight – including overweight and obese as defined by BMI – may also independently influence the success of treatment. “Our data suggest that if you have RA, it’s important to take the medications that your doctor has prescribed. If you smoke, you need to stop. And if you’re carrying extra weight, not only is that placing a greater demand on already vulnerable joints, it may also be making your RA treatment less effective,” Dr. Bartlett said.
These lifestyle modifications can be challenging for some people with RA, she said in the interview. Clinicians can help by considering lifestyle behaviors that lead to chronic diseases and poorer outcomes in addition to their more traditional view of diagnosis and treatment, she said, adding that patients and clinicians should know that even a small amount of weight loss can improve health and may improve response to therapy.
Well controlled clinical trials will be needed to better understand the benefits of weight control and smoking cessation on response to RA treatment. Also, why women who smoke and are overweight are at more of a disadvantage than their male counterparts is unknown. “As we begin putting these pieces together, we may learn valuable information that helps us to better control and ultimately cure RA,” said Dr. Bartlett.
The researchers had no conflicts of interest to declare.
The video associated with this article is no longer available on this site. Please view all of our videos on the MDedge YouTube channel
LONDON – Tobacco use and excess weight can make it harder to achieve sustained remission in the treatment of early rheumatoid arthritis, according to findings from more than 1,000 patients in the Canadian Early Arthritis Cohort (CATCH) multicenter, prospective study.
Aggressive treatment that starts soon after diagnosis of rheumatoid arthritis (RA) is important for the absence of disease activity, which is the hallmark of sustained remission. But the reality is a success rate of less than 50% in the first 3 years with physical deterioration continuing thereafter. “Excess weight and smoking are two risk factors for developing RA. We were interested in seeing if they might also affect how well people responded to treatment,” said Susan Bartlett, Ph.D., a clinical psychologist at McGill University in Montreal.
At the annual European Congress of Rheumatology, Dr. Bartlett and colleagues reported on a cohort of 1,008 early RA patients who were enrolled in the Canadian Early Arthritis Cohort (CATCH) multicenter, prospective study and followed from around the time of diagnosis through the first 3 years of treatment to estimate the time it took until they achieved sustained remission, defined as having a 28-joint Disease Activity Score less than 2.6 for two consecutive visits.
Mean age of the patients (72% female, 81% white) was early 50s. Overall, 30% of females and 47% of males were overweight, one-third of both genders were obese, and 15%-20% smoked. Treatment at entry included methotrexate in mono- or combination therapy in about three-quarters of the patients, with steroids used in about half and biologics used sparingly.
The proportion of patients in sustained remission was 38% at 3 years, with a median time to remission of 11.3 months. “That finding wasn’t surprising because that is generally what is found in most studies of early RA. However, when we looked more closely at who was and wasn’t achieving remission, we found that people who smoked and those who were overweight or obese were much less likely than their nonsmoking, normal-weight peers to be in sustained remission,” Dr. Bartlett said in a pre-congress interview.
After adjustment for factors that could affect response to treatment – including age, race, disability status, pain, and early medications used – smoking (P = .046) and excess weight (P = .003) were associated with a poorer likelihood of achieving sustained remission. While more men than women were overweight or obese, the effects of weight and smoking appeared to be more problematic for women (P = .02).
An average nonsmoking male with a healthy body mass index (BMI; 25 kg/m2 or less) had about a 41% probability of achieving sustained remission within 3 years, compared with 15% for an obese male smoker. A nonsmoking female with a healthy BMI had a 27% probability of achieving sustained remission within 3 years, compared with 10% for an obese female smoker. Probabilities of sustained remission were also lower for overweight men and women, Dr. Bartlett reported.
Smoking and obesity have already been linked with an increased likelihood of developing RA, which in turn increases the risk of cardiovascular disease and premature death. The latest data suggest that both smoking and extra weight – including overweight and obese as defined by BMI – may also independently influence the success of treatment. “Our data suggest that if you have RA, it’s important to take the medications that your doctor has prescribed. If you smoke, you need to stop. And if you’re carrying extra weight, not only is that placing a greater demand on already vulnerable joints, it may also be making your RA treatment less effective,” Dr. Bartlett said.
These lifestyle modifications can be challenging for some people with RA, she said in the interview. Clinicians can help by considering lifestyle behaviors that lead to chronic diseases and poorer outcomes in addition to their more traditional view of diagnosis and treatment, she said, adding that patients and clinicians should know that even a small amount of weight loss can improve health and may improve response to therapy.
Well controlled clinical trials will be needed to better understand the benefits of weight control and smoking cessation on response to RA treatment. Also, why women who smoke and are overweight are at more of a disadvantage than their male counterparts is unknown. “As we begin putting these pieces together, we may learn valuable information that helps us to better control and ultimately cure RA,” said Dr. Bartlett.
The researchers had no conflicts of interest to declare.
The video associated with this article is no longer available on this site. Please view all of our videos on the MDedge YouTube channel
LONDON – Tobacco use and excess weight can make it harder to achieve sustained remission in the treatment of early rheumatoid arthritis, according to findings from more than 1,000 patients in the Canadian Early Arthritis Cohort (CATCH) multicenter, prospective study.
Aggressive treatment that starts soon after diagnosis of rheumatoid arthritis (RA) is important for the absence of disease activity, which is the hallmark of sustained remission. But the reality is a success rate of less than 50% in the first 3 years with physical deterioration continuing thereafter. “Excess weight and smoking are two risk factors for developing RA. We were interested in seeing if they might also affect how well people responded to treatment,” said Susan Bartlett, Ph.D., a clinical psychologist at McGill University in Montreal.
At the annual European Congress of Rheumatology, Dr. Bartlett and colleagues reported on a cohort of 1,008 early RA patients who were enrolled in the Canadian Early Arthritis Cohort (CATCH) multicenter, prospective study and followed from around the time of diagnosis through the first 3 years of treatment to estimate the time it took until they achieved sustained remission, defined as having a 28-joint Disease Activity Score less than 2.6 for two consecutive visits.
Mean age of the patients (72% female, 81% white) was early 50s. Overall, 30% of females and 47% of males were overweight, one-third of both genders were obese, and 15%-20% smoked. Treatment at entry included methotrexate in mono- or combination therapy in about three-quarters of the patients, with steroids used in about half and biologics used sparingly.
The proportion of patients in sustained remission was 38% at 3 years, with a median time to remission of 11.3 months. “That finding wasn’t surprising because that is generally what is found in most studies of early RA. However, when we looked more closely at who was and wasn’t achieving remission, we found that people who smoked and those who were overweight or obese were much less likely than their nonsmoking, normal-weight peers to be in sustained remission,” Dr. Bartlett said in a pre-congress interview.
After adjustment for factors that could affect response to treatment – including age, race, disability status, pain, and early medications used – smoking (P = .046) and excess weight (P = .003) were associated with a poorer likelihood of achieving sustained remission. While more men than women were overweight or obese, the effects of weight and smoking appeared to be more problematic for women (P = .02).
An average nonsmoking male with a healthy body mass index (BMI; 25 kg/m2 or less) had about a 41% probability of achieving sustained remission within 3 years, compared with 15% for an obese male smoker. A nonsmoking female with a healthy BMI had a 27% probability of achieving sustained remission within 3 years, compared with 10% for an obese female smoker. Probabilities of sustained remission were also lower for overweight men and women, Dr. Bartlett reported.
Smoking and obesity have already been linked with an increased likelihood of developing RA, which in turn increases the risk of cardiovascular disease and premature death. The latest data suggest that both smoking and extra weight – including overweight and obese as defined by BMI – may also independently influence the success of treatment. “Our data suggest that if you have RA, it’s important to take the medications that your doctor has prescribed. If you smoke, you need to stop. And if you’re carrying extra weight, not only is that placing a greater demand on already vulnerable joints, it may also be making your RA treatment less effective,” Dr. Bartlett said.
These lifestyle modifications can be challenging for some people with RA, she said in the interview. Clinicians can help by considering lifestyle behaviors that lead to chronic diseases and poorer outcomes in addition to their more traditional view of diagnosis and treatment, she said, adding that patients and clinicians should know that even a small amount of weight loss can improve health and may improve response to therapy.
Well controlled clinical trials will be needed to better understand the benefits of weight control and smoking cessation on response to RA treatment. Also, why women who smoke and are overweight are at more of a disadvantage than their male counterparts is unknown. “As we begin putting these pieces together, we may learn valuable information that helps us to better control and ultimately cure RA,” said Dr. Bartlett.
The researchers had no conflicts of interest to declare.
The video associated with this article is no longer available on this site. Please view all of our videos on the MDedge YouTube channel
AT THE EULAR CONGRESS 2016
Key clinical point: The latest data suggest that both smoking and extra weight – including overweight and obese as defined by BMI – may independently influence the success of treatment for rheumatoid arthritis.
Major finding: A nonsmoking female with a healthy BMI had a 27% probability of achieving sustained remission within 3 years, compared with 10% for an obese female smoker.
Data source: A prospective study of 1,008 patients with early RA in the prospective, multicenter CATCH study.
Disclosures: The researchers had no conflicts of interest to declare.
Teriflunomide effective for faster-advancing MS
NATIONAL HARBOR, MD. – The approved once-daily oral immunomodulator teriflunomide is effective in reducing relapse, and worsening of relapsing forms, of multiple sclerosis (RMS) in patients with faster-advancing disease.
The findings from a post hoc pooled subgroup analysis of patients in the TEMSO and TOWER randomized, double-blind, placebo-controlled phase III trials presented as a poster at the annual meeting of the Consortium of Multiple Sclerosis Centers expand the efficacy of teriflunomide to a broad spectrum of patients with RMS.
The pooled analysis examined the outcomes of treatment with two doses of teriflunomide in all patients (n = 2,257) and in a subgroup of patients with faster-advancing MS (n = 1,184) according to disease severity using a baseline Multiple Sclerosis Severity Score (MSSS) of 5 as a cutoff. The group with faster-advancing disease included 405 who received placebo, 396 treated with teriflunomide 7 mg, and 383 treated with teriflunomide 14 mg.
The 1,184 patients with faster-advancing MS were comparable to the other 1,073 patients in terms of age, sex, MS subtype, and number of relapses in the prior year. Baseline characteristics of the faster-advancing MS patients treated with placebo and both teriflunomide doses were comparable.
Relative to the placebo group, the annualized relapse rate in patients with faster-advancing MS was significantly reduced by both doses of teriflunomide, with a relative risk reduction of 30.3% for the 7-mg dose (P = .0002) and 37.5% for the 14-mg dose (P less than .0001). Time to first relapse was also reduced by teriflunomide 14 mg and 7 mg, with reductions in relapse risk of 43.2% (P less than .0001) and 28.8% (P = .0014), respectively, compared with placebo.
The higher dose of teriflunomide was effective in reducing the worsening of disability at greater than or equal to 12 weeks and 24 weeks, with reductions, compared with placebo, of 40.3% (P = .0076) and 46.1% (P = .0110), respectively.
The results were consistent with data reported in the individual TEMSO and TOWER trials, and prior analyses of the pooled data.
“These results indicate that teriflunomide is not only effective for MS patients with relatively mild disease, but also with rather more aggressive disease. Teriflunomide, a well-tolerated and convenient once-daily tablet, is a very reasonable option for this population,” said the presenter, Dr. Aaron E. Miller of the Icahn School of Medicine at Mount Sinai, New York.
Teriflunomide is approved for relapsing-remitting MS in many countries, including the United States and the European Union. The TEMSO and TOWER trials were pivotal in securing approval. The trials had a similar design, except for the absence of magnetic resonance imaging evaluation in TOWER.
The study was funded by Sanofi Genzyme. Dr. Miller disclosed consulting fees from Accordant Health Services, Acorda Therapeutics, Alkermes, Biogen, EMD Serono, Genentech, Genzyme, GlaxoSmithKline, Mallinckrodt Paharmaceuticals/Questor, Novartis, Roche, and Teva, and grant/research support from Biogen, Genentech, Novartis, Questor, Roche, and Sanofi.
NATIONAL HARBOR, MD. – The approved once-daily oral immunomodulator teriflunomide is effective in reducing relapse, and worsening of relapsing forms, of multiple sclerosis (RMS) in patients with faster-advancing disease.
The findings from a post hoc pooled subgroup analysis of patients in the TEMSO and TOWER randomized, double-blind, placebo-controlled phase III trials presented as a poster at the annual meeting of the Consortium of Multiple Sclerosis Centers expand the efficacy of teriflunomide to a broad spectrum of patients with RMS.
The pooled analysis examined the outcomes of treatment with two doses of teriflunomide in all patients (n = 2,257) and in a subgroup of patients with faster-advancing MS (n = 1,184) according to disease severity using a baseline Multiple Sclerosis Severity Score (MSSS) of 5 as a cutoff. The group with faster-advancing disease included 405 who received placebo, 396 treated with teriflunomide 7 mg, and 383 treated with teriflunomide 14 mg.
The 1,184 patients with faster-advancing MS were comparable to the other 1,073 patients in terms of age, sex, MS subtype, and number of relapses in the prior year. Baseline characteristics of the faster-advancing MS patients treated with placebo and both teriflunomide doses were comparable.
Relative to the placebo group, the annualized relapse rate in patients with faster-advancing MS was significantly reduced by both doses of teriflunomide, with a relative risk reduction of 30.3% for the 7-mg dose (P = .0002) and 37.5% for the 14-mg dose (P less than .0001). Time to first relapse was also reduced by teriflunomide 14 mg and 7 mg, with reductions in relapse risk of 43.2% (P less than .0001) and 28.8% (P = .0014), respectively, compared with placebo.
The higher dose of teriflunomide was effective in reducing the worsening of disability at greater than or equal to 12 weeks and 24 weeks, with reductions, compared with placebo, of 40.3% (P = .0076) and 46.1% (P = .0110), respectively.
The results were consistent with data reported in the individual TEMSO and TOWER trials, and prior analyses of the pooled data.
“These results indicate that teriflunomide is not only effective for MS patients with relatively mild disease, but also with rather more aggressive disease. Teriflunomide, a well-tolerated and convenient once-daily tablet, is a very reasonable option for this population,” said the presenter, Dr. Aaron E. Miller of the Icahn School of Medicine at Mount Sinai, New York.
Teriflunomide is approved for relapsing-remitting MS in many countries, including the United States and the European Union. The TEMSO and TOWER trials were pivotal in securing approval. The trials had a similar design, except for the absence of magnetic resonance imaging evaluation in TOWER.
The study was funded by Sanofi Genzyme. Dr. Miller disclosed consulting fees from Accordant Health Services, Acorda Therapeutics, Alkermes, Biogen, EMD Serono, Genentech, Genzyme, GlaxoSmithKline, Mallinckrodt Paharmaceuticals/Questor, Novartis, Roche, and Teva, and grant/research support from Biogen, Genentech, Novartis, Questor, Roche, and Sanofi.
NATIONAL HARBOR, MD. – The approved once-daily oral immunomodulator teriflunomide is effective in reducing relapse, and worsening of relapsing forms, of multiple sclerosis (RMS) in patients with faster-advancing disease.
The findings from a post hoc pooled subgroup analysis of patients in the TEMSO and TOWER randomized, double-blind, placebo-controlled phase III trials presented as a poster at the annual meeting of the Consortium of Multiple Sclerosis Centers expand the efficacy of teriflunomide to a broad spectrum of patients with RMS.
The pooled analysis examined the outcomes of treatment with two doses of teriflunomide in all patients (n = 2,257) and in a subgroup of patients with faster-advancing MS (n = 1,184) according to disease severity using a baseline Multiple Sclerosis Severity Score (MSSS) of 5 as a cutoff. The group with faster-advancing disease included 405 who received placebo, 396 treated with teriflunomide 7 mg, and 383 treated with teriflunomide 14 mg.
The 1,184 patients with faster-advancing MS were comparable to the other 1,073 patients in terms of age, sex, MS subtype, and number of relapses in the prior year. Baseline characteristics of the faster-advancing MS patients treated with placebo and both teriflunomide doses were comparable.
Relative to the placebo group, the annualized relapse rate in patients with faster-advancing MS was significantly reduced by both doses of teriflunomide, with a relative risk reduction of 30.3% for the 7-mg dose (P = .0002) and 37.5% for the 14-mg dose (P less than .0001). Time to first relapse was also reduced by teriflunomide 14 mg and 7 mg, with reductions in relapse risk of 43.2% (P less than .0001) and 28.8% (P = .0014), respectively, compared with placebo.
The higher dose of teriflunomide was effective in reducing the worsening of disability at greater than or equal to 12 weeks and 24 weeks, with reductions, compared with placebo, of 40.3% (P = .0076) and 46.1% (P = .0110), respectively.
The results were consistent with data reported in the individual TEMSO and TOWER trials, and prior analyses of the pooled data.
“These results indicate that teriflunomide is not only effective for MS patients with relatively mild disease, but also with rather more aggressive disease. Teriflunomide, a well-tolerated and convenient once-daily tablet, is a very reasonable option for this population,” said the presenter, Dr. Aaron E. Miller of the Icahn School of Medicine at Mount Sinai, New York.
Teriflunomide is approved for relapsing-remitting MS in many countries, including the United States and the European Union. The TEMSO and TOWER trials were pivotal in securing approval. The trials had a similar design, except for the absence of magnetic resonance imaging evaluation in TOWER.
The study was funded by Sanofi Genzyme. Dr. Miller disclosed consulting fees from Accordant Health Services, Acorda Therapeutics, Alkermes, Biogen, EMD Serono, Genentech, Genzyme, GlaxoSmithKline, Mallinckrodt Paharmaceuticals/Questor, Novartis, Roche, and Teva, and grant/research support from Biogen, Genentech, Novartis, Questor, Roche, and Sanofi.
AT THE CMSC ANNUAL MEETING
Key clinical point: Teriflunomide is effective for patients with faster-advancing MS.
Major finding: Teriflunomide at 14 mg and 7 mg reduced the annualized relapse rate of patients with fast-advancing relapsing-remitting MS (MSSS greater than 5) by 43.2% and 28.8%, respectively, compared with placebo.
Data source: A pooled analysis of patients from the TEMSO and TOWER randomized, double-blind, placebo-controlled phase III trials.
Disclosures: The study was funded by Sanofi Genzyme. Dr. Miller disclosed consulting fees from Accordant Health Services, Acorda Therapeutics, Alkermes, Biogen, EMD Serono, Genentech, Genzyme, GlaxoSmithKline, Mallinckrodt Paharmaceuticals/Questor, Novartis, Roche, and Teva, and grant/research support from Biogen, Genentech, Novartis, Questor, Roche, and Sanofi.
Smoking causes worse outcome in MS
NATIONAL HARBOR, MD. – Follow-up of a cohort of patients in the United Kingdom has demonstrated associations between smoking and a higher risk of development of multiple sclerosis (MS), progression of MS-related disability, higher risk of premature death, and shortened life expectancy.
The findings highlight the need for clinical trials of the effects of quitting smoking and provide data that will be useful in the development of effective intervention strategies.
Dr. Cris S. Constantinescu of the University of Nottingham (England) discussed findings from the Nottingham University Hospitals MS Clinics database at the annual meeting of the Consortium of Multiple Sclerosis Centers.
“Although smokers had higher levels of comorbid conditions, it appeared that the influence of smoking is independent of the presence of comorbid conditions. Those who gave up smoking could do as well as nonsmokers,” said Dr. Constantinescu.
While ample epidemiologic evidence indicates that smoking is a driver of the development and progression of MS, there are few hard data. To gain insight, the researchers analyzed data on over 1,200 MS patients throughout England who were followed up beginning in the mid-1990s. About 60% of the patients had relapsing-remitting MS. The duration of MS and smoking were both about 20 years. About 60% of men and 50% of women were current smokers.
Regular smokers were 64% more likely to develop MS than were nonsmokers. Having ever smoked carried a 44% increased risk of MS (both P less than .001). MS patients who grew up in a household where one parent smoked were 50% more likely to become regular smokers. The risk climbed to 85% if both parents were smokers.
No association was evident between smoking and the development of primary progressive MS, but current smokers were almost 2.5 times more likely to develop secondary progressive MS. Smoking correlated with more severe MS disability, compared with nonsmokers. Ex-smokers had risks similar to those of nonsmokers of developing secondary MS and in the level of disease severity.
Every year a person refrained from smoking decreased the risk of severe disability by 5%. Current and ex-smokers displayed increased psychological and physical detriments of MS.
In a subgroup of 923 patients, of whom 80 died, current smokers were almost 3 times and 1.5 times more likely to die, compared with never smokers and ex-smokers, respectively, and were twice as likely to die as were people without MS in the UK general population.
The findings have prompted studies into major aspects of smoking, such as the age when smoking begins, the success of various smoking cessation programs, and development of interventions. Some of the data discussed at the meeting were published in 2013 (Brain;136:2298-2304) and some are part of a manuscript in preparation.
NATIONAL HARBOR, MD. – Follow-up of a cohort of patients in the United Kingdom has demonstrated associations between smoking and a higher risk of development of multiple sclerosis (MS), progression of MS-related disability, higher risk of premature death, and shortened life expectancy.
The findings highlight the need for clinical trials of the effects of quitting smoking and provide data that will be useful in the development of effective intervention strategies.
Dr. Cris S. Constantinescu of the University of Nottingham (England) discussed findings from the Nottingham University Hospitals MS Clinics database at the annual meeting of the Consortium of Multiple Sclerosis Centers.
“Although smokers had higher levels of comorbid conditions, it appeared that the influence of smoking is independent of the presence of comorbid conditions. Those who gave up smoking could do as well as nonsmokers,” said Dr. Constantinescu.
While ample epidemiologic evidence indicates that smoking is a driver of the development and progression of MS, there are few hard data. To gain insight, the researchers analyzed data on over 1,200 MS patients throughout England who were followed up beginning in the mid-1990s. About 60% of the patients had relapsing-remitting MS. The duration of MS and smoking were both about 20 years. About 60% of men and 50% of women were current smokers.
Regular smokers were 64% more likely to develop MS than were nonsmokers. Having ever smoked carried a 44% increased risk of MS (both P less than .001). MS patients who grew up in a household where one parent smoked were 50% more likely to become regular smokers. The risk climbed to 85% if both parents were smokers.
No association was evident between smoking and the development of primary progressive MS, but current smokers were almost 2.5 times more likely to develop secondary progressive MS. Smoking correlated with more severe MS disability, compared with nonsmokers. Ex-smokers had risks similar to those of nonsmokers of developing secondary MS and in the level of disease severity.
Every year a person refrained from smoking decreased the risk of severe disability by 5%. Current and ex-smokers displayed increased psychological and physical detriments of MS.
In a subgroup of 923 patients, of whom 80 died, current smokers were almost 3 times and 1.5 times more likely to die, compared with never smokers and ex-smokers, respectively, and were twice as likely to die as were people without MS in the UK general population.
The findings have prompted studies into major aspects of smoking, such as the age when smoking begins, the success of various smoking cessation programs, and development of interventions. Some of the data discussed at the meeting were published in 2013 (Brain;136:2298-2304) and some are part of a manuscript in preparation.
NATIONAL HARBOR, MD. – Follow-up of a cohort of patients in the United Kingdom has demonstrated associations between smoking and a higher risk of development of multiple sclerosis (MS), progression of MS-related disability, higher risk of premature death, and shortened life expectancy.
The findings highlight the need for clinical trials of the effects of quitting smoking and provide data that will be useful in the development of effective intervention strategies.
Dr. Cris S. Constantinescu of the University of Nottingham (England) discussed findings from the Nottingham University Hospitals MS Clinics database at the annual meeting of the Consortium of Multiple Sclerosis Centers.
“Although smokers had higher levels of comorbid conditions, it appeared that the influence of smoking is independent of the presence of comorbid conditions. Those who gave up smoking could do as well as nonsmokers,” said Dr. Constantinescu.
While ample epidemiologic evidence indicates that smoking is a driver of the development and progression of MS, there are few hard data. To gain insight, the researchers analyzed data on over 1,200 MS patients throughout England who were followed up beginning in the mid-1990s. About 60% of the patients had relapsing-remitting MS. The duration of MS and smoking were both about 20 years. About 60% of men and 50% of women were current smokers.
Regular smokers were 64% more likely to develop MS than were nonsmokers. Having ever smoked carried a 44% increased risk of MS (both P less than .001). MS patients who grew up in a household where one parent smoked were 50% more likely to become regular smokers. The risk climbed to 85% if both parents were smokers.
No association was evident between smoking and the development of primary progressive MS, but current smokers were almost 2.5 times more likely to develop secondary progressive MS. Smoking correlated with more severe MS disability, compared with nonsmokers. Ex-smokers had risks similar to those of nonsmokers of developing secondary MS and in the level of disease severity.
Every year a person refrained from smoking decreased the risk of severe disability by 5%. Current and ex-smokers displayed increased psychological and physical detriments of MS.
In a subgroup of 923 patients, of whom 80 died, current smokers were almost 3 times and 1.5 times more likely to die, compared with never smokers and ex-smokers, respectively, and were twice as likely to die as were people without MS in the UK general population.
The findings have prompted studies into major aspects of smoking, such as the age when smoking begins, the success of various smoking cessation programs, and development of interventions. Some of the data discussed at the meeting were published in 2013 (Brain;136:2298-2304) and some are part of a manuscript in preparation.
AT THE CMSC ANNUAL MEETING
Key clinical point: Smoking increases the risk of developing multiple sclerosis and worsens the severity of the disease.
Major finding: Regular smokers are 64% more likely to develop MS than are nonsmokers.
Data source: Nottingham MS Clinic Study.
Disclosures: Dr. Constantinescu reported having no financial disclosures.
MS imaging guidelines from North American group offer standardized protocols
NATIONAL HARBOR, MD. – Updated imaging protocols for patients with multiple sclerosis from a panel of North American neurology and radiology experts promise to improve the accuracy of diagnosis and monitoring.
The guidelines emphasize the use of three-dimensional MRI to provide complete coverage of the brain, monitoring for progressive multifocal leukoencephalopathy (PML), and optical orbit MRI for severe optic neuritis.
Key clinical guideline changes include more specific timing of brain MRI when monitoring patients receiving disease modifying therapy, timing of brain MRI to monitor for PML, updated evidence of the value of MRI changes in determining the effectiveness of treatment, and the inclusion of radiologic isolated syndrome.
Dr. Anthony Traboulsee of the University of British Columbia, Vancouver, discussed the latest MRI guidelines at the annual meeting of the Consortium of Multiple Sclerosis Centers.
“MS is a life-long disorder and MRI is one of the best ways to monitor for new lesions that can be occurring in the absence of new symptoms (clinically silent disease activity). These lesions accumulate and lead to future disability. In order to accurately determine if changes are occurring, we need MRIs that are similar in quality over time regardless of where the patient had an MRI performed. A standardized MRI protocol provides this quality of data,” Dr. Traboulsee said.
The guidelines, published in the American Journal of Neuroradiology (Am J Neuroradiol. 2015 Nov 12. doi: 10.3174/ajnr.A4539), recommend a first MRI as soon as a physician suspects MS in a patient, with subsequent MRIs typically done annually to determine whether the disease is stable or progressive, which could prompt a change in treatment. “Breakthrough activity on MRI that is occurring while on treatment can lead to future disability and is an opportunity to consider switching therapy,” Dr. Traboulsee said.
More frequent scans could be appropriate for patients with a more aggressive and active disease course, and when treatment has been changed. More frequent monitoring and diffusion-weighted imaging is also recommended for patients taking natalizumab (Tysabri), since they are at high risk for PML.
Another goal of the guidelines is to describe how best to use MRI to support an early diagnosis of MS and to help avoid misdiagnosis. Early treatment depends on an early diagnosis. “We find the biggest impact of our disease-modifying therapy is in the first decade. The goal is to prevent new injury and optimize brain health through treatment and lifestyle,” said Dr. Traboulsee.
MRI scans alone should not be diagnostic. Clinical information is also needed, such as numbness or problems with balance. Abnormal brain MRIs occur in about 5% of people without MS and white spots in the brain that are unrelated to MS can naturally develop as people age.
If clinically isolated syndrome is suspected, a cervical cord MRI should be done along with a brain MRI. Use of a gadolinium contrast agent is recommended to better determine disease activity and speed diagnosis.
Dr. Traboulsee received grant support from Biogen, Chugai, Hoffman la Roche, and Sanofi Genzyme. He reported being a steering committee member for Hoffman la Roche and has been a consultant to Biogen, Chugai, EMD Serono, Hoffman la Roche, MedImmune, Sanofi Genzyme, and Teva Neuroscience.
NATIONAL HARBOR, MD. – Updated imaging protocols for patients with multiple sclerosis from a panel of North American neurology and radiology experts promise to improve the accuracy of diagnosis and monitoring.
The guidelines emphasize the use of three-dimensional MRI to provide complete coverage of the brain, monitoring for progressive multifocal leukoencephalopathy (PML), and optical orbit MRI for severe optic neuritis.
Key clinical guideline changes include more specific timing of brain MRI when monitoring patients receiving disease modifying therapy, timing of brain MRI to monitor for PML, updated evidence of the value of MRI changes in determining the effectiveness of treatment, and the inclusion of radiologic isolated syndrome.
Dr. Anthony Traboulsee of the University of British Columbia, Vancouver, discussed the latest MRI guidelines at the annual meeting of the Consortium of Multiple Sclerosis Centers.
“MS is a life-long disorder and MRI is one of the best ways to monitor for new lesions that can be occurring in the absence of new symptoms (clinically silent disease activity). These lesions accumulate and lead to future disability. In order to accurately determine if changes are occurring, we need MRIs that are similar in quality over time regardless of where the patient had an MRI performed. A standardized MRI protocol provides this quality of data,” Dr. Traboulsee said.
The guidelines, published in the American Journal of Neuroradiology (Am J Neuroradiol. 2015 Nov 12. doi: 10.3174/ajnr.A4539), recommend a first MRI as soon as a physician suspects MS in a patient, with subsequent MRIs typically done annually to determine whether the disease is stable or progressive, which could prompt a change in treatment. “Breakthrough activity on MRI that is occurring while on treatment can lead to future disability and is an opportunity to consider switching therapy,” Dr. Traboulsee said.
More frequent scans could be appropriate for patients with a more aggressive and active disease course, and when treatment has been changed. More frequent monitoring and diffusion-weighted imaging is also recommended for patients taking natalizumab (Tysabri), since they are at high risk for PML.
Another goal of the guidelines is to describe how best to use MRI to support an early diagnosis of MS and to help avoid misdiagnosis. Early treatment depends on an early diagnosis. “We find the biggest impact of our disease-modifying therapy is in the first decade. The goal is to prevent new injury and optimize brain health through treatment and lifestyle,” said Dr. Traboulsee.
MRI scans alone should not be diagnostic. Clinical information is also needed, such as numbness or problems with balance. Abnormal brain MRIs occur in about 5% of people without MS and white spots in the brain that are unrelated to MS can naturally develop as people age.
If clinically isolated syndrome is suspected, a cervical cord MRI should be done along with a brain MRI. Use of a gadolinium contrast agent is recommended to better determine disease activity and speed diagnosis.
Dr. Traboulsee received grant support from Biogen, Chugai, Hoffman la Roche, and Sanofi Genzyme. He reported being a steering committee member for Hoffman la Roche and has been a consultant to Biogen, Chugai, EMD Serono, Hoffman la Roche, MedImmune, Sanofi Genzyme, and Teva Neuroscience.
NATIONAL HARBOR, MD. – Updated imaging protocols for patients with multiple sclerosis from a panel of North American neurology and radiology experts promise to improve the accuracy of diagnosis and monitoring.
The guidelines emphasize the use of three-dimensional MRI to provide complete coverage of the brain, monitoring for progressive multifocal leukoencephalopathy (PML), and optical orbit MRI for severe optic neuritis.
Key clinical guideline changes include more specific timing of brain MRI when monitoring patients receiving disease modifying therapy, timing of brain MRI to monitor for PML, updated evidence of the value of MRI changes in determining the effectiveness of treatment, and the inclusion of radiologic isolated syndrome.
Dr. Anthony Traboulsee of the University of British Columbia, Vancouver, discussed the latest MRI guidelines at the annual meeting of the Consortium of Multiple Sclerosis Centers.
“MS is a life-long disorder and MRI is one of the best ways to monitor for new lesions that can be occurring in the absence of new symptoms (clinically silent disease activity). These lesions accumulate and lead to future disability. In order to accurately determine if changes are occurring, we need MRIs that are similar in quality over time regardless of where the patient had an MRI performed. A standardized MRI protocol provides this quality of data,” Dr. Traboulsee said.
The guidelines, published in the American Journal of Neuroradiology (Am J Neuroradiol. 2015 Nov 12. doi: 10.3174/ajnr.A4539), recommend a first MRI as soon as a physician suspects MS in a patient, with subsequent MRIs typically done annually to determine whether the disease is stable or progressive, which could prompt a change in treatment. “Breakthrough activity on MRI that is occurring while on treatment can lead to future disability and is an opportunity to consider switching therapy,” Dr. Traboulsee said.
More frequent scans could be appropriate for patients with a more aggressive and active disease course, and when treatment has been changed. More frequent monitoring and diffusion-weighted imaging is also recommended for patients taking natalizumab (Tysabri), since they are at high risk for PML.
Another goal of the guidelines is to describe how best to use MRI to support an early diagnosis of MS and to help avoid misdiagnosis. Early treatment depends on an early diagnosis. “We find the biggest impact of our disease-modifying therapy is in the first decade. The goal is to prevent new injury and optimize brain health through treatment and lifestyle,” said Dr. Traboulsee.
MRI scans alone should not be diagnostic. Clinical information is also needed, such as numbness or problems with balance. Abnormal brain MRIs occur in about 5% of people without MS and white spots in the brain that are unrelated to MS can naturally develop as people age.
If clinically isolated syndrome is suspected, a cervical cord MRI should be done along with a brain MRI. Use of a gadolinium contrast agent is recommended to better determine disease activity and speed diagnosis.
Dr. Traboulsee received grant support from Biogen, Chugai, Hoffman la Roche, and Sanofi Genzyme. He reported being a steering committee member for Hoffman la Roche and has been a consultant to Biogen, Chugai, EMD Serono, Hoffman la Roche, MedImmune, Sanofi Genzyme, and Teva Neuroscience.
AT THE CMSC ANNUAL MEETING
Beloranib reduces weight in Prader-Willi syndrome, but concerns remain
BOSTON – Prader-Willi syndrome patients lost more than 4% of their body weight over a 26-week period with the investigative antiobesity drug beloranib, compared with a similar gain in patients taking placebo, in the phase III bestPWS trial.
Hunger-related behavior was also reduced, along with total and LDL cholesterol levels and other cardiometabolic risk factors, lead investigator Dr. Merlin G. Butler reported at the annual meeting of the Endocrinology Society.
However, because of concerns over venous thromboembolic events in patients on beloranib, the trial was halted before all patients reached 26 weeks of treatment.
“bestPWS is the first phase III clinical trial to show statistically and clinically significant weight loss and improvement in hyperphagia-related behaviors in PWS [Prader-Willi syndrome] patients. The reduction in hyperphagia-related behaviors in the beloranib-treated groups represents a clinically meaningful benefit to patients,” Dr. Butler said.
Beloranib is a novel, first-in-class injectable molecule that works by inhibiting MetAP2, an enzyme that modulates the activity of cellular processes that are important in the control of metabolism. Its benefits in preclinical studies and a phase II trial in reducing body weight and decreasing hyperplasia fueled optimism regarding the therapeutic value in Prader-Willi syndrome, the most common genetic cause of morbid obesity. Although rare, Prader-Willi syndrome is life threatening and life limiting, with most of those affected dying before the age of 50.
“There are currently no treatment options for the intractable obesity and hyperphagia in Prader-Willi syndrome,” said Dr. Butler of the University of Kansas Medical Center, Kansas City, Kan.
In the bestPWS trial, 107 patients were randomized 2:1 to receive twice-weekly subcutaneous injections of beloranib or placebo for 26 weeks: 36 received 1.8 mg and 37 received 2.4 mg of beloranib, and 34 received placebo. Seventy-four patients completed the treatment. The coprimary efficacy endpoints were improvement in hyperphagia-related behaviors and reduction in body weight. Secondary endpoints included improvements in total body fat mass, lipids, and, as markers of cardiometabolic risk, total cholesterol and LDL cholesterol levels.
Baseline demographic and clinical characteristics were comparable in the three trial arms, with an average age of 20 years, average body mass index of 40 kg/m2, average body weight of 100 kg, average fat mass of 51 kg, and average Hyperphagia Questionnaire for Clinical Trials (HQ-CT) total score of 16.9.
Patients in the placebo arm displayed an increase in body weight of about 4% over the 26-week trial. In contrast, patients treated with beloranib lost weight (4.1% and 5.3% in the 1.8-mg and 2.4-mg arms, respectively; both P less than .0001, compared with placebo). The weight loss in the two treatment arms did not differ significantly. Beloranib was also associated with improvements in body composition, total cholesterol, and LDL cholesterol, high-sensitivity C-reactive protein, leptin, and adiponectin, compared with placebo.
Both beloranib doses also appreciably reduced hunger-associated behaviors, with reductions of 6.7-7.4 points with beloranib, compared with a reduction of 0.4 with placebo.
Adverse events most commonly included injection-site bruising, aggression, and hyperphagia; they were generally mild and transient.
There were five serious adverse events. Those that occurred in the treatment arms were psychiatric disorders, which are common background comorbidities in Prader-Willi patients, and so are not necessarily treatment related. In the bestPWS trial, two deep vein thrombosis events and two fatal pulmonary embolism events occurred. After the first pulmonary embolism death, the trial was discontinued; at that point, 27 randomized patients had not completed the full 26-week course.
Overall, 11 venous thrombotic events, including pulmonary embolism, deep vein thrombosis, and superficial thrombophlebitis, have occurred in the roughly 400 patients who have so far received beloranib in the course of its development, including the adverse events in the bestPWS trial. None of these events has occurred in those receiving placebo.
Even in light of these sobering events, Dr. Butler said he remains optimistic. “In addition to the reduction in body weight and decrease in excessive eating behaviors previously reported from this study, [these data] demonstrate important reductions in cardiometabolic risk factors and further support a strong rationale for continued evaluation of beloranib as a potential treatment for Prader-Willi syndrome,” said Dr. Butler.
Beloranib is currently on clinical hold while the potential for a prothrombotic effect of beloranib and a heightened risk for Prader-Willi syndrome patients are assessed, Dr. Butler said.
Dr. Butler disclosed study funding by Zafgen, the manufacturer of Beloranib.
BOSTON – Prader-Willi syndrome patients lost more than 4% of their body weight over a 26-week period with the investigative antiobesity drug beloranib, compared with a similar gain in patients taking placebo, in the phase III bestPWS trial.
Hunger-related behavior was also reduced, along with total and LDL cholesterol levels and other cardiometabolic risk factors, lead investigator Dr. Merlin G. Butler reported at the annual meeting of the Endocrinology Society.
However, because of concerns over venous thromboembolic events in patients on beloranib, the trial was halted before all patients reached 26 weeks of treatment.
“bestPWS is the first phase III clinical trial to show statistically and clinically significant weight loss and improvement in hyperphagia-related behaviors in PWS [Prader-Willi syndrome] patients. The reduction in hyperphagia-related behaviors in the beloranib-treated groups represents a clinically meaningful benefit to patients,” Dr. Butler said.
Beloranib is a novel, first-in-class injectable molecule that works by inhibiting MetAP2, an enzyme that modulates the activity of cellular processes that are important in the control of metabolism. Its benefits in preclinical studies and a phase II trial in reducing body weight and decreasing hyperplasia fueled optimism regarding the therapeutic value in Prader-Willi syndrome, the most common genetic cause of morbid obesity. Although rare, Prader-Willi syndrome is life threatening and life limiting, with most of those affected dying before the age of 50.
“There are currently no treatment options for the intractable obesity and hyperphagia in Prader-Willi syndrome,” said Dr. Butler of the University of Kansas Medical Center, Kansas City, Kan.
In the bestPWS trial, 107 patients were randomized 2:1 to receive twice-weekly subcutaneous injections of beloranib or placebo for 26 weeks: 36 received 1.8 mg and 37 received 2.4 mg of beloranib, and 34 received placebo. Seventy-four patients completed the treatment. The coprimary efficacy endpoints were improvement in hyperphagia-related behaviors and reduction in body weight. Secondary endpoints included improvements in total body fat mass, lipids, and, as markers of cardiometabolic risk, total cholesterol and LDL cholesterol levels.
Baseline demographic and clinical characteristics were comparable in the three trial arms, with an average age of 20 years, average body mass index of 40 kg/m2, average body weight of 100 kg, average fat mass of 51 kg, and average Hyperphagia Questionnaire for Clinical Trials (HQ-CT) total score of 16.9.
Patients in the placebo arm displayed an increase in body weight of about 4% over the 26-week trial. In contrast, patients treated with beloranib lost weight (4.1% and 5.3% in the 1.8-mg and 2.4-mg arms, respectively; both P less than .0001, compared with placebo). The weight loss in the two treatment arms did not differ significantly. Beloranib was also associated with improvements in body composition, total cholesterol, and LDL cholesterol, high-sensitivity C-reactive protein, leptin, and adiponectin, compared with placebo.
Both beloranib doses also appreciably reduced hunger-associated behaviors, with reductions of 6.7-7.4 points with beloranib, compared with a reduction of 0.4 with placebo.
Adverse events most commonly included injection-site bruising, aggression, and hyperphagia; they were generally mild and transient.
There were five serious adverse events. Those that occurred in the treatment arms were psychiatric disorders, which are common background comorbidities in Prader-Willi patients, and so are not necessarily treatment related. In the bestPWS trial, two deep vein thrombosis events and two fatal pulmonary embolism events occurred. After the first pulmonary embolism death, the trial was discontinued; at that point, 27 randomized patients had not completed the full 26-week course.
Overall, 11 venous thrombotic events, including pulmonary embolism, deep vein thrombosis, and superficial thrombophlebitis, have occurred in the roughly 400 patients who have so far received beloranib in the course of its development, including the adverse events in the bestPWS trial. None of these events has occurred in those receiving placebo.
Even in light of these sobering events, Dr. Butler said he remains optimistic. “In addition to the reduction in body weight and decrease in excessive eating behaviors previously reported from this study, [these data] demonstrate important reductions in cardiometabolic risk factors and further support a strong rationale for continued evaluation of beloranib as a potential treatment for Prader-Willi syndrome,” said Dr. Butler.
Beloranib is currently on clinical hold while the potential for a prothrombotic effect of beloranib and a heightened risk for Prader-Willi syndrome patients are assessed, Dr. Butler said.
Dr. Butler disclosed study funding by Zafgen, the manufacturer of Beloranib.
BOSTON – Prader-Willi syndrome patients lost more than 4% of their body weight over a 26-week period with the investigative antiobesity drug beloranib, compared with a similar gain in patients taking placebo, in the phase III bestPWS trial.
Hunger-related behavior was also reduced, along with total and LDL cholesterol levels and other cardiometabolic risk factors, lead investigator Dr. Merlin G. Butler reported at the annual meeting of the Endocrinology Society.
However, because of concerns over venous thromboembolic events in patients on beloranib, the trial was halted before all patients reached 26 weeks of treatment.
“bestPWS is the first phase III clinical trial to show statistically and clinically significant weight loss and improvement in hyperphagia-related behaviors in PWS [Prader-Willi syndrome] patients. The reduction in hyperphagia-related behaviors in the beloranib-treated groups represents a clinically meaningful benefit to patients,” Dr. Butler said.
Beloranib is a novel, first-in-class injectable molecule that works by inhibiting MetAP2, an enzyme that modulates the activity of cellular processes that are important in the control of metabolism. Its benefits in preclinical studies and a phase II trial in reducing body weight and decreasing hyperplasia fueled optimism regarding the therapeutic value in Prader-Willi syndrome, the most common genetic cause of morbid obesity. Although rare, Prader-Willi syndrome is life threatening and life limiting, with most of those affected dying before the age of 50.
“There are currently no treatment options for the intractable obesity and hyperphagia in Prader-Willi syndrome,” said Dr. Butler of the University of Kansas Medical Center, Kansas City, Kan.
In the bestPWS trial, 107 patients were randomized 2:1 to receive twice-weekly subcutaneous injections of beloranib or placebo for 26 weeks: 36 received 1.8 mg and 37 received 2.4 mg of beloranib, and 34 received placebo. Seventy-four patients completed the treatment. The coprimary efficacy endpoints were improvement in hyperphagia-related behaviors and reduction in body weight. Secondary endpoints included improvements in total body fat mass, lipids, and, as markers of cardiometabolic risk, total cholesterol and LDL cholesterol levels.
Baseline demographic and clinical characteristics were comparable in the three trial arms, with an average age of 20 years, average body mass index of 40 kg/m2, average body weight of 100 kg, average fat mass of 51 kg, and average Hyperphagia Questionnaire for Clinical Trials (HQ-CT) total score of 16.9.
Patients in the placebo arm displayed an increase in body weight of about 4% over the 26-week trial. In contrast, patients treated with beloranib lost weight (4.1% and 5.3% in the 1.8-mg and 2.4-mg arms, respectively; both P less than .0001, compared with placebo). The weight loss in the two treatment arms did not differ significantly. Beloranib was also associated with improvements in body composition, total cholesterol, and LDL cholesterol, high-sensitivity C-reactive protein, leptin, and adiponectin, compared with placebo.
Both beloranib doses also appreciably reduced hunger-associated behaviors, with reductions of 6.7-7.4 points with beloranib, compared with a reduction of 0.4 with placebo.
Adverse events most commonly included injection-site bruising, aggression, and hyperphagia; they were generally mild and transient.
There were five serious adverse events. Those that occurred in the treatment arms were psychiatric disorders, which are common background comorbidities in Prader-Willi patients, and so are not necessarily treatment related. In the bestPWS trial, two deep vein thrombosis events and two fatal pulmonary embolism events occurred. After the first pulmonary embolism death, the trial was discontinued; at that point, 27 randomized patients had not completed the full 26-week course.
Overall, 11 venous thrombotic events, including pulmonary embolism, deep vein thrombosis, and superficial thrombophlebitis, have occurred in the roughly 400 patients who have so far received beloranib in the course of its development, including the adverse events in the bestPWS trial. None of these events has occurred in those receiving placebo.
Even in light of these sobering events, Dr. Butler said he remains optimistic. “In addition to the reduction in body weight and decrease in excessive eating behaviors previously reported from this study, [these data] demonstrate important reductions in cardiometabolic risk factors and further support a strong rationale for continued evaluation of beloranib as a potential treatment for Prader-Willi syndrome,” said Dr. Butler.
Beloranib is currently on clinical hold while the potential for a prothrombotic effect of beloranib and a heightened risk for Prader-Willi syndrome patients are assessed, Dr. Butler said.
Dr. Butler disclosed study funding by Zafgen, the manufacturer of Beloranib.
AT ENDO 2016
Key clinical point: Beloranib produces significant weight loss in patients with Prader-Willi syndrome, but concerns have arisen about associated pulmonary emboli.
Major finding: Patients with Prader-Willi syndrome on beloranib lost 4.1%-5.3% of their body weight, compared with a gain of 4% in placebo patients; however, 11 venous thrombotic events occurred in the beloranib patients.
Data source: bestPWS, a randomized, double-blind, placebo-controlled trial in 107 patients.
Disclosures: Dr. Butler disclosed study funding by Zafgen, the manufacturer of Beloranib.
VIDEO: Proposed revision of medullary thyroid cancer staging improves risk-stratification analysis
BOSTON – An analysis of data from medullary thyroid cancer patients that partitioned the patients into groups with similar overall survival has spurred a rethink of the current American Joint Committee on Cancer (AJCC) staging system.
The results from researchers at Duke University, Durham, N.C., presented at the annual meeting of the Endocrine Society by Dr. Mohamed Abdelgadir Adam, are timely, as the AJCC has embarked on a reconsideration of the staging of cancers, including medullary thyroid cancer (MTC), as part revisions for the eighth edition of the staging system.
“The existing AJCC staging system for MTC appears to be less than optimal in discriminating the risk of mortality among disease stage groups,” said Dr. Adam, who discussed the findings in a video interview.
MTC, a neuroendocrine tumor that affects C cells of the thyroid, comprises 3%-5% of all cases of thyroid cancer and it can be a more aggressive disease than differentiated thyroid cancer. Yet the current AJCC MTC staging system has been extrapolated from differentiated thyroid cancer data.
“We sought to evaluate how well the current AJCC seventh edition stage groupings predict survival for patients with MTC, to suggest a possible staging revision to sharpen estimates of prognosis,” said Dr. Adam.
The researchers utilized the National Cancer Data Base, representing over 70% of incident cancer cases in the United States.
MTC patients who underwent thyroid surgery from 1998 to 2012 were identified. Patients with missing values for pathologic T, N, or M were excluded. The primary outcome in the 3,315 patients was survival.
The researchers used a form of decision-tree analysis called recursive partitioning. In general, recursive partitioning is able to classify a population by splitting subjects into subgroups, each of which is homogeneous based on the particular outcome. In this study, the subgroup allocations were based on T, N, and M stages, with the outcome being overall survival. Kaplan-Meier and adjusted survival analyses enabled survival differences among the four subgroups (groups I, II, III and IV) to be explored.
The four groups were distinct in terms of survival time and allowed more accurate risk stratification. In particular, groups I and II were markedly better distinguished from one another than is the case with the current staging system. Survival differences across the stages were more distinct with the newly created T, N, and M groupings, compared with the current AJCC staging system.
After adjustment, survival differences across TNM groups were more distinct with the newly created TNM groupings (compared to subgroup I, hazard ratio of 3.06 for subgroup II; HR, 6.79 for III; and HR, 17.03 for IV), compared with the current AJCC staging (compared to stage I, HR, 1.45 for stage II; HR, 2.17 for III; and HR, 5.33 for IV).
“The AJCC is reevaluating all staging schemas, including MTC. The current AJCC staging system could be improved with the newly identified TNM groupings suggested here for more accurate patient risk stratification and possibly treatment selection,” said Dr. Adam.
Dr. Adam had no disclosures.
The video associated with this article is no longer available on this site. Please view all of our videos on the MDedge YouTube channel
BOSTON – An analysis of data from medullary thyroid cancer patients that partitioned the patients into groups with similar overall survival has spurred a rethink of the current American Joint Committee on Cancer (AJCC) staging system.
The results from researchers at Duke University, Durham, N.C., presented at the annual meeting of the Endocrine Society by Dr. Mohamed Abdelgadir Adam, are timely, as the AJCC has embarked on a reconsideration of the staging of cancers, including medullary thyroid cancer (MTC), as part revisions for the eighth edition of the staging system.
“The existing AJCC staging system for MTC appears to be less than optimal in discriminating the risk of mortality among disease stage groups,” said Dr. Adam, who discussed the findings in a video interview.
MTC, a neuroendocrine tumor that affects C cells of the thyroid, comprises 3%-5% of all cases of thyroid cancer and it can be a more aggressive disease than differentiated thyroid cancer. Yet the current AJCC MTC staging system has been extrapolated from differentiated thyroid cancer data.
“We sought to evaluate how well the current AJCC seventh edition stage groupings predict survival for patients with MTC, to suggest a possible staging revision to sharpen estimates of prognosis,” said Dr. Adam.
The researchers utilized the National Cancer Data Base, representing over 70% of incident cancer cases in the United States.
MTC patients who underwent thyroid surgery from 1998 to 2012 were identified. Patients with missing values for pathologic T, N, or M were excluded. The primary outcome in the 3,315 patients was survival.
The researchers used a form of decision-tree analysis called recursive partitioning. In general, recursive partitioning is able to classify a population by splitting subjects into subgroups, each of which is homogeneous based on the particular outcome. In this study, the subgroup allocations were based on T, N, and M stages, with the outcome being overall survival. Kaplan-Meier and adjusted survival analyses enabled survival differences among the four subgroups (groups I, II, III and IV) to be explored.
The four groups were distinct in terms of survival time and allowed more accurate risk stratification. In particular, groups I and II were markedly better distinguished from one another than is the case with the current staging system. Survival differences across the stages were more distinct with the newly created T, N, and M groupings, compared with the current AJCC staging system.
After adjustment, survival differences across TNM groups were more distinct with the newly created TNM groupings (compared to subgroup I, hazard ratio of 3.06 for subgroup II; HR, 6.79 for III; and HR, 17.03 for IV), compared with the current AJCC staging (compared to stage I, HR, 1.45 for stage II; HR, 2.17 for III; and HR, 5.33 for IV).
“The AJCC is reevaluating all staging schemas, including MTC. The current AJCC staging system could be improved with the newly identified TNM groupings suggested here for more accurate patient risk stratification and possibly treatment selection,” said Dr. Adam.
Dr. Adam had no disclosures.
The video associated with this article is no longer available on this site. Please view all of our videos on the MDedge YouTube channel
BOSTON – An analysis of data from medullary thyroid cancer patients that partitioned the patients into groups with similar overall survival has spurred a rethink of the current American Joint Committee on Cancer (AJCC) staging system.
The results from researchers at Duke University, Durham, N.C., presented at the annual meeting of the Endocrine Society by Dr. Mohamed Abdelgadir Adam, are timely, as the AJCC has embarked on a reconsideration of the staging of cancers, including medullary thyroid cancer (MTC), as part revisions for the eighth edition of the staging system.
“The existing AJCC staging system for MTC appears to be less than optimal in discriminating the risk of mortality among disease stage groups,” said Dr. Adam, who discussed the findings in a video interview.
MTC, a neuroendocrine tumor that affects C cells of the thyroid, comprises 3%-5% of all cases of thyroid cancer and it can be a more aggressive disease than differentiated thyroid cancer. Yet the current AJCC MTC staging system has been extrapolated from differentiated thyroid cancer data.
“We sought to evaluate how well the current AJCC seventh edition stage groupings predict survival for patients with MTC, to suggest a possible staging revision to sharpen estimates of prognosis,” said Dr. Adam.
The researchers utilized the National Cancer Data Base, representing over 70% of incident cancer cases in the United States.
MTC patients who underwent thyroid surgery from 1998 to 2012 were identified. Patients with missing values for pathologic T, N, or M were excluded. The primary outcome in the 3,315 patients was survival.
The researchers used a form of decision-tree analysis called recursive partitioning. In general, recursive partitioning is able to classify a population by splitting subjects into subgroups, each of which is homogeneous based on the particular outcome. In this study, the subgroup allocations were based on T, N, and M stages, with the outcome being overall survival. Kaplan-Meier and adjusted survival analyses enabled survival differences among the four subgroups (groups I, II, III and IV) to be explored.
The four groups were distinct in terms of survival time and allowed more accurate risk stratification. In particular, groups I and II were markedly better distinguished from one another than is the case with the current staging system. Survival differences across the stages were more distinct with the newly created T, N, and M groupings, compared with the current AJCC staging system.
After adjustment, survival differences across TNM groups were more distinct with the newly created TNM groupings (compared to subgroup I, hazard ratio of 3.06 for subgroup II; HR, 6.79 for III; and HR, 17.03 for IV), compared with the current AJCC staging (compared to stage I, HR, 1.45 for stage II; HR, 2.17 for III; and HR, 5.33 for IV).
“The AJCC is reevaluating all staging schemas, including MTC. The current AJCC staging system could be improved with the newly identified TNM groupings suggested here for more accurate patient risk stratification and possibly treatment selection,” said Dr. Adam.
Dr. Adam had no disclosures.
The video associated with this article is no longer available on this site. Please view all of our videos on the MDedge YouTube channel
Key clinical point: A proposed revision of the AJCC thyroid cancer staging system improves risk stratification analysis.
Major finding: In the proposed staging system, compared to subgroup I, hazard ratio for survival was 3.06 for subgroup II; HR, 6.79 for III; and HR, 17.03 for IV, compared with the current AJCC staging of HR, 1.45 for stage II; HR, 2.17 for III; and HR, 5.33 for IV.
Data source: Data from 3,315 patients with medullary thyroid cancer was drawn from the National Cancer Database.
Disclosures: Dr. Adam had no disclosures.
VIDEO: Weight cycling common following weight loss in obese individuals
BOSTON – Examination of weight-loss patterns in over 177,000 people has revealed that, regardless of the initial 6-month weight loss, after 2 years the majority of patients become “cyclers,” with periods of weight gain and loss rather than maintenance of the initial weight loss.
“One-third of American adults are obese. In 2010, the cost of obesity and obesity-related comorbidities in the United States was estimated to be $315.8 billion. Achieving and maintaining weight loss has proven to be difficult,” said Joanna Huang, PharmD, senior manager of health economics and outcomes research at Novo Nordisk, Plainsboro, N.J., and lead investigator of the study presented at the annual meeting of the Endocrine Society.
The study examined the electronic records of about 178,000 obese patients whose weight loss had been by deliberate intent and not due to illness. The subjects were allocated into four groups based on the extent of weight loss in terms of body mass index (BMI) over 6 months: Those who remained stable and lost less than 5% (n = 151,902), those who lost 5%-10% (modest loss; n = 16,637), those who lost 10%-15% (moderate loss; n = 4,035), and those who lost in excess of 15% (high loss; n = 5,945).
The subjects who were at least 18 years of age at baseline (mean age 54-58 years), had at least one BMI measurement that was indicative of obesity (greater than or equal to 30 kg/m2), with at least four BMI determinations done over at least 5 years. Subjects were mostly white (about 66% in all four groups) and mostly from the southern United States.
Regardless of the amount that the participants lost in the first 6 months, regain of 50% of more of body weight was common in the modest weight-loss group (40%) and moderate weight-loss group (36%), while only 19% of those in the high weight-loss group cycled back up in weight, reported study presenter Maral DerSarkissian, PhD, of the Analysis Group in Boston.
More than 73% and about 70% of those in the moderate and modest weight-loss group, respectively, experienced weight cycling within 2 years. In the stable and high weight-loss groups, the situation was somewhat more optimistic, with about 60% of participants cycling in weight within 2 years. Total regain of lost weight occurred in about 23%, 16%, and 7% of the modest, moderate, and high weight-loss group, respectively.
“Weight loss maintenance, even in the moderate and high weight-loss groups, is very difficult to achieve,” said Dr. Huang.
Interventions that seek to maintain the weight conventionally are directed at dietary changes. But, according to Dr. DerSarkissian, “these modifications alone might not be enough to achieve and maintain weight loss.”
Pharmacotherapy is another weight-loss option. The data indicated that only 2% of the participants were receiving weight-loss pharmacotherapy. Whether this figure is accurate is an open question, according to Dr. Huang, since a lot of the data were compiled from physicians’ notes. Since clinicians may not record weight-loss advice offered to their patients, the data base may well not reflect lifestyle interventions, including pharmacotherapy.
In addition, since the data captured only primary outpatient care, whether or not a patient ever had bariatric surgery was unknown. Other unrecorded factors that can influence weight over time included comorbidities, use of medications, diet changes, and changes in physical activity.
The data points to a multifactor approach to weight loss that includes counseling, positive reinforcement, dietary advice, pharmacotherapy where appropriate, and, in some cases, bariatric surgery.
“Successful and sustained clinically meaningful weight loss requires chronic and effective weight management strategies,” said Dr. Huang.
Dr. Huang is an employee of Novo Nordisk and Dr. DerSarkissian is a researcher for Novo Nordisk.
The video associated with this article is no longer available on this site. Please view all of our videos on the MDedge YouTube channel
BOSTON – Examination of weight-loss patterns in over 177,000 people has revealed that, regardless of the initial 6-month weight loss, after 2 years the majority of patients become “cyclers,” with periods of weight gain and loss rather than maintenance of the initial weight loss.
“One-third of American adults are obese. In 2010, the cost of obesity and obesity-related comorbidities in the United States was estimated to be $315.8 billion. Achieving and maintaining weight loss has proven to be difficult,” said Joanna Huang, PharmD, senior manager of health economics and outcomes research at Novo Nordisk, Plainsboro, N.J., and lead investigator of the study presented at the annual meeting of the Endocrine Society.
The study examined the electronic records of about 178,000 obese patients whose weight loss had been by deliberate intent and not due to illness. The subjects were allocated into four groups based on the extent of weight loss in terms of body mass index (BMI) over 6 months: Those who remained stable and lost less than 5% (n = 151,902), those who lost 5%-10% (modest loss; n = 16,637), those who lost 10%-15% (moderate loss; n = 4,035), and those who lost in excess of 15% (high loss; n = 5,945).
The subjects who were at least 18 years of age at baseline (mean age 54-58 years), had at least one BMI measurement that was indicative of obesity (greater than or equal to 30 kg/m2), with at least four BMI determinations done over at least 5 years. Subjects were mostly white (about 66% in all four groups) and mostly from the southern United States.
Regardless of the amount that the participants lost in the first 6 months, regain of 50% of more of body weight was common in the modest weight-loss group (40%) and moderate weight-loss group (36%), while only 19% of those in the high weight-loss group cycled back up in weight, reported study presenter Maral DerSarkissian, PhD, of the Analysis Group in Boston.
More than 73% and about 70% of those in the moderate and modest weight-loss group, respectively, experienced weight cycling within 2 years. In the stable and high weight-loss groups, the situation was somewhat more optimistic, with about 60% of participants cycling in weight within 2 years. Total regain of lost weight occurred in about 23%, 16%, and 7% of the modest, moderate, and high weight-loss group, respectively.
“Weight loss maintenance, even in the moderate and high weight-loss groups, is very difficult to achieve,” said Dr. Huang.
Interventions that seek to maintain the weight conventionally are directed at dietary changes. But, according to Dr. DerSarkissian, “these modifications alone might not be enough to achieve and maintain weight loss.”
Pharmacotherapy is another weight-loss option. The data indicated that only 2% of the participants were receiving weight-loss pharmacotherapy. Whether this figure is accurate is an open question, according to Dr. Huang, since a lot of the data were compiled from physicians’ notes. Since clinicians may not record weight-loss advice offered to their patients, the data base may well not reflect lifestyle interventions, including pharmacotherapy.
In addition, since the data captured only primary outpatient care, whether or not a patient ever had bariatric surgery was unknown. Other unrecorded factors that can influence weight over time included comorbidities, use of medications, diet changes, and changes in physical activity.
The data points to a multifactor approach to weight loss that includes counseling, positive reinforcement, dietary advice, pharmacotherapy where appropriate, and, in some cases, bariatric surgery.
“Successful and sustained clinically meaningful weight loss requires chronic and effective weight management strategies,” said Dr. Huang.
Dr. Huang is an employee of Novo Nordisk and Dr. DerSarkissian is a researcher for Novo Nordisk.
The video associated with this article is no longer available on this site. Please view all of our videos on the MDedge YouTube channel
BOSTON – Examination of weight-loss patterns in over 177,000 people has revealed that, regardless of the initial 6-month weight loss, after 2 years the majority of patients become “cyclers,” with periods of weight gain and loss rather than maintenance of the initial weight loss.
“One-third of American adults are obese. In 2010, the cost of obesity and obesity-related comorbidities in the United States was estimated to be $315.8 billion. Achieving and maintaining weight loss has proven to be difficult,” said Joanna Huang, PharmD, senior manager of health economics and outcomes research at Novo Nordisk, Plainsboro, N.J., and lead investigator of the study presented at the annual meeting of the Endocrine Society.
The study examined the electronic records of about 178,000 obese patients whose weight loss had been by deliberate intent and not due to illness. The subjects were allocated into four groups based on the extent of weight loss in terms of body mass index (BMI) over 6 months: Those who remained stable and lost less than 5% (n = 151,902), those who lost 5%-10% (modest loss; n = 16,637), those who lost 10%-15% (moderate loss; n = 4,035), and those who lost in excess of 15% (high loss; n = 5,945).
The subjects who were at least 18 years of age at baseline (mean age 54-58 years), had at least one BMI measurement that was indicative of obesity (greater than or equal to 30 kg/m2), with at least four BMI determinations done over at least 5 years. Subjects were mostly white (about 66% in all four groups) and mostly from the southern United States.
Regardless of the amount that the participants lost in the first 6 months, regain of 50% of more of body weight was common in the modest weight-loss group (40%) and moderate weight-loss group (36%), while only 19% of those in the high weight-loss group cycled back up in weight, reported study presenter Maral DerSarkissian, PhD, of the Analysis Group in Boston.
More than 73% and about 70% of those in the moderate and modest weight-loss group, respectively, experienced weight cycling within 2 years. In the stable and high weight-loss groups, the situation was somewhat more optimistic, with about 60% of participants cycling in weight within 2 years. Total regain of lost weight occurred in about 23%, 16%, and 7% of the modest, moderate, and high weight-loss group, respectively.
“Weight loss maintenance, even in the moderate and high weight-loss groups, is very difficult to achieve,” said Dr. Huang.
Interventions that seek to maintain the weight conventionally are directed at dietary changes. But, according to Dr. DerSarkissian, “these modifications alone might not be enough to achieve and maintain weight loss.”
Pharmacotherapy is another weight-loss option. The data indicated that only 2% of the participants were receiving weight-loss pharmacotherapy. Whether this figure is accurate is an open question, according to Dr. Huang, since a lot of the data were compiled from physicians’ notes. Since clinicians may not record weight-loss advice offered to their patients, the data base may well not reflect lifestyle interventions, including pharmacotherapy.
In addition, since the data captured only primary outpatient care, whether or not a patient ever had bariatric surgery was unknown. Other unrecorded factors that can influence weight over time included comorbidities, use of medications, diet changes, and changes in physical activity.
The data points to a multifactor approach to weight loss that includes counseling, positive reinforcement, dietary advice, pharmacotherapy where appropriate, and, in some cases, bariatric surgery.
“Successful and sustained clinically meaningful weight loss requires chronic and effective weight management strategies,” said Dr. Huang.
Dr. Huang is an employee of Novo Nordisk and Dr. DerSarkissian is a researcher for Novo Nordisk.
The video associated with this article is no longer available on this site. Please view all of our videos on the MDedge YouTube channel
AT ENDO 2016
Key clinical point: Most patients who lose modest or moderate amounts of weight experience periods of both gain and loss within 2 years.
Major finding: More than 73% and about 70% of those in the moderate and modest weight-loss group, respectively, experienced weight cycling within 2 years. Total regain of lost weight occurred in about 23%, 16%, and 7% of the modest, moderate, and high weight-loss group, respectively.
Data source: Electronic records of 177,743 obese patients whose weight loss had been by deliberate intent and not due to illness.
Disclosures: Dr. Huang is an employee of Novo Nordisk and Dr. DerSarkissian is a researcher for Novo Nordisk.
T2DM Patients on Combination Therapy Benefit in Switch From Sitagliptin to Liraglutide
BOSTON – Switching from sitagliptin to liraglutide, in combination with metformin, improved control of hypoglycemia and resulted in greater weight loss in patients with type 2 diabetes, reported Dr. Maximo Maislos at the annual meeting of the Endocrine Society.
Results of a randomized, double-blind, double-dummy, active-controlled 26-week trial have indicated that liraglutide can be used as an add-on to metformin for patients with type 2 diabetes who have remained hyperglycemic.
“Switching from sitagliptin to liraglutide resulted in superior [glycated hemoglobin] and body weight reductions, compared with continued sitagliptin treatment,” said Dr. Maximo Maislos of Ben-Gurion University, Beer-Sheva, Israel.
The LIRA-SWITCH trial (Efficacy and Safety of Switching From Sitagliptin to Liraglutide in Subjects With Type 2 Diabetes Not Achieving Adequate Glycaemic Control on Sitagliptin and Metformin) involved 407 patients. The majority (60%) were male; mean age was 56 years and mean body mass index was 32 kg/m2. The subjects had all been treated with sitagliptin (100 mg/day) and metformin (greater than or equal to 1,500 mg/day or a maximum tolerated dose greater than or equal to 1,000 mg/day) for at least 90 days. Hyperglycemia had not been well controlled, with a mean hemoglobin A1C (HbAIC) level of 8.3% (67 mmol/mol). The mean duration of type 2 diabetes was 8 years.
Subjects were randomized to continued sitagliptin along with metformin (n = 204) or liraglutide (1.8 mg daily) along with metformin (n = 203).
After 26 weeks of treatment, reduction in HbAIC was significantly greater in the liraglutide arm than in the sitagliptin arm (1.14% vs. 0.54%; estimated treatment difference [ETD], –0.61%; 95% confidence interval, –0.82 to –0.40; P less than .0001). Those receiving liraglutide had statistically significantly greater weight loss, compared with those who continued on sitagliptin.
The less than 7% and less than or equal to 6.5% target levels of HbAIC were achieved by 50.6% and 29.5%, respectively, of patients in the liraglutide arm. These percentages were significantly higher than the respective 26.9% and 9.9% of patients in the sitagliptin arm (P less than .0001 for both). Fasting plasma glucose levels were significantly reduced with liraglutide treatment while decreases in systolic and diastolic blood pressure were similar in the two study arms.
Adverse events (AEs) occurred more often in the liraglutide group than in the sitagliptin group (68.8% vs. 56.9%). Thirteen patients receiving liraglutide discontinued treatment, compared with five in the sitagliptin arm. The most common AEs in the liraglutide group were gastrointestinal disorders, principally nausea (21.8% with liraglutide vs. 7.8% with sitagliptin) and diarrhea (16.3% with liraglutide vs. 9.3% with sitagliptin), followed by decreased appetite (8.9% vs. 3.4%, respectively). These AEs tended to subside within the first few weeks of treatment.
Serious AEs occurred in eight patients in both arms. Rescue medication was needed for 30 patients receiving sitagliptin and 11 patients receiving liraglutide. No cases of pancreatitis were reported. In the sitagliptin group, one subject each developed bladder cancer and squamous cell carcinoma. Nocturnal hypoglycemia did not develop in either trial arm.
Funding was provided by liraglutide maker Novo Nordisk. Dr. Maislos had no disclosures.
BOSTON – Switching from sitagliptin to liraglutide, in combination with metformin, improved control of hypoglycemia and resulted in greater weight loss in patients with type 2 diabetes, reported Dr. Maximo Maislos at the annual meeting of the Endocrine Society.
Results of a randomized, double-blind, double-dummy, active-controlled 26-week trial have indicated that liraglutide can be used as an add-on to metformin for patients with type 2 diabetes who have remained hyperglycemic.
“Switching from sitagliptin to liraglutide resulted in superior [glycated hemoglobin] and body weight reductions, compared with continued sitagliptin treatment,” said Dr. Maximo Maislos of Ben-Gurion University, Beer-Sheva, Israel.
The LIRA-SWITCH trial (Efficacy and Safety of Switching From Sitagliptin to Liraglutide in Subjects With Type 2 Diabetes Not Achieving Adequate Glycaemic Control on Sitagliptin and Metformin) involved 407 patients. The majority (60%) were male; mean age was 56 years and mean body mass index was 32 kg/m2. The subjects had all been treated with sitagliptin (100 mg/day) and metformin (greater than or equal to 1,500 mg/day or a maximum tolerated dose greater than or equal to 1,000 mg/day) for at least 90 days. Hyperglycemia had not been well controlled, with a mean hemoglobin A1C (HbAIC) level of 8.3% (67 mmol/mol). The mean duration of type 2 diabetes was 8 years.
Subjects were randomized to continued sitagliptin along with metformin (n = 204) or liraglutide (1.8 mg daily) along with metformin (n = 203).
After 26 weeks of treatment, reduction in HbAIC was significantly greater in the liraglutide arm than in the sitagliptin arm (1.14% vs. 0.54%; estimated treatment difference [ETD], –0.61%; 95% confidence interval, –0.82 to –0.40; P less than .0001). Those receiving liraglutide had statistically significantly greater weight loss, compared with those who continued on sitagliptin.
The less than 7% and less than or equal to 6.5% target levels of HbAIC were achieved by 50.6% and 29.5%, respectively, of patients in the liraglutide arm. These percentages were significantly higher than the respective 26.9% and 9.9% of patients in the sitagliptin arm (P less than .0001 for both). Fasting plasma glucose levels were significantly reduced with liraglutide treatment while decreases in systolic and diastolic blood pressure were similar in the two study arms.
Adverse events (AEs) occurred more often in the liraglutide group than in the sitagliptin group (68.8% vs. 56.9%). Thirteen patients receiving liraglutide discontinued treatment, compared with five in the sitagliptin arm. The most common AEs in the liraglutide group were gastrointestinal disorders, principally nausea (21.8% with liraglutide vs. 7.8% with sitagliptin) and diarrhea (16.3% with liraglutide vs. 9.3% with sitagliptin), followed by decreased appetite (8.9% vs. 3.4%, respectively). These AEs tended to subside within the first few weeks of treatment.
Serious AEs occurred in eight patients in both arms. Rescue medication was needed for 30 patients receiving sitagliptin and 11 patients receiving liraglutide. No cases of pancreatitis were reported. In the sitagliptin group, one subject each developed bladder cancer and squamous cell carcinoma. Nocturnal hypoglycemia did not develop in either trial arm.
Funding was provided by liraglutide maker Novo Nordisk. Dr. Maislos had no disclosures.
BOSTON – Switching from sitagliptin to liraglutide, in combination with metformin, improved control of hypoglycemia and resulted in greater weight loss in patients with type 2 diabetes, reported Dr. Maximo Maislos at the annual meeting of the Endocrine Society.
Results of a randomized, double-blind, double-dummy, active-controlled 26-week trial have indicated that liraglutide can be used as an add-on to metformin for patients with type 2 diabetes who have remained hyperglycemic.
“Switching from sitagliptin to liraglutide resulted in superior [glycated hemoglobin] and body weight reductions, compared with continued sitagliptin treatment,” said Dr. Maximo Maislos of Ben-Gurion University, Beer-Sheva, Israel.
The LIRA-SWITCH trial (Efficacy and Safety of Switching From Sitagliptin to Liraglutide in Subjects With Type 2 Diabetes Not Achieving Adequate Glycaemic Control on Sitagliptin and Metformin) involved 407 patients. The majority (60%) were male; mean age was 56 years and mean body mass index was 32 kg/m2. The subjects had all been treated with sitagliptin (100 mg/day) and metformin (greater than or equal to 1,500 mg/day or a maximum tolerated dose greater than or equal to 1,000 mg/day) for at least 90 days. Hyperglycemia had not been well controlled, with a mean hemoglobin A1C (HbAIC) level of 8.3% (67 mmol/mol). The mean duration of type 2 diabetes was 8 years.
Subjects were randomized to continued sitagliptin along with metformin (n = 204) or liraglutide (1.8 mg daily) along with metformin (n = 203).
After 26 weeks of treatment, reduction in HbAIC was significantly greater in the liraglutide arm than in the sitagliptin arm (1.14% vs. 0.54%; estimated treatment difference [ETD], –0.61%; 95% confidence interval, –0.82 to –0.40; P less than .0001). Those receiving liraglutide had statistically significantly greater weight loss, compared with those who continued on sitagliptin.
The less than 7% and less than or equal to 6.5% target levels of HbAIC were achieved by 50.6% and 29.5%, respectively, of patients in the liraglutide arm. These percentages were significantly higher than the respective 26.9% and 9.9% of patients in the sitagliptin arm (P less than .0001 for both). Fasting plasma glucose levels were significantly reduced with liraglutide treatment while decreases in systolic and diastolic blood pressure were similar in the two study arms.
Adverse events (AEs) occurred more often in the liraglutide group than in the sitagliptin group (68.8% vs. 56.9%). Thirteen patients receiving liraglutide discontinued treatment, compared with five in the sitagliptin arm. The most common AEs in the liraglutide group were gastrointestinal disorders, principally nausea (21.8% with liraglutide vs. 7.8% with sitagliptin) and diarrhea (16.3% with liraglutide vs. 9.3% with sitagliptin), followed by decreased appetite (8.9% vs. 3.4%, respectively). These AEs tended to subside within the first few weeks of treatment.
Serious AEs occurred in eight patients in both arms. Rescue medication was needed for 30 patients receiving sitagliptin and 11 patients receiving liraglutide. No cases of pancreatitis were reported. In the sitagliptin group, one subject each developed bladder cancer and squamous cell carcinoma. Nocturnal hypoglycemia did not develop in either trial arm.
Funding was provided by liraglutide maker Novo Nordisk. Dr. Maislos had no disclosures.
AT ENDO 2016
T2D patients on combination therapy benefit in switch from sitagliptin to liraglutide
BOSTON – Switching from sitagliptin to liraglutide, in combination with metformin, improved control of hypoglycemia and resulted in greater weight loss in patients with type 2 diabetes, reported Dr. Maximo Maislos at the annual meeting of the Endocrine Society.
Results of a randomized, double-blind, double-dummy, active-controlled 26-week trial have indicated that liraglutide can be used as an add-on to metformin for patients with type 2 diabetes who have remained hyperglycemic.
“Switching from sitagliptin to liraglutide resulted in superior [glycated hemoglobin] and body weight reductions, compared with continued sitagliptin treatment,” said Dr. Maximo Maislos of Ben-Gurion University, Beer-Sheva, Israel.
The LIRA-SWITCH trial (Efficacy and Safety of Switching From Sitagliptin to Liraglutide in Subjects With Type 2 Diabetes Not Achieving Adequate Glycaemic Control on Sitagliptin and Metformin) involved 407 patients. The majority (60%) were male; mean age was 56 years and mean body mass index was 32 kg/m2. The subjects had all been treated with sitagliptin (100 mg/day) and metformin (greater than or equal to 1,500 mg/day or a maximum tolerated dose greater than or equal to 1,000 mg/day) for at least 90 days. Hyperglycemia had not been well controlled, with a mean hemoglobin A1C (HbAIC) level of 8.3% (67 mmol/mol). The mean duration of type 2 diabetes was 8 years.
Subjects were randomized to continued sitagliptin along with metformin (n = 204) or liraglutide (1.8 mg daily) along with metformin (n = 203).
After 26 weeks of treatment, reduction in HbAIC was significantly greater in the liraglutide arm than in the sitagliptin arm (1.14% vs. 0.54%; estimated treatment difference [ETD], –0.61%; 95% confidence interval, –0.82 to –0.40; P less than .0001). Those receiving liraglutide had statistically significantly greater weight loss, compared with those who continued on sitagliptin.
The less than 7% and less than or equal to 6.5% target levels of HbAIC were achieved by 50.6% and 29.5%, respectively, of patients in the liraglutide arm. These percentages were significantly higher than the respective 26.9% and 9.9% of patients in the sitagliptin arm (P less than .0001 for both). Fasting plasma glucose levels were significantly reduced with liraglutide treatment while decreases in systolic and diastolic blood pressure were similar in the two study arms.
Adverse events (AEs) occurred more often in the liraglutide group than in the sitagliptin group (68.8% vs. 56.9%). Thirteen patients receiving liraglutide discontinued treatment, compared with five in the sitagliptin arm. The most common AEs in the liraglutide group were gastrointestinal disorders, principally nausea (21.8% with liraglutide vs. 7.8% with sitagliptin) and diarrhea (16.3% with liraglutide vs. 9.3% with sitagliptin), followed by decreased appetite (8.9% vs. 3.4%, respectively). These AEs tended to subside within the first few weeks of treatment.
Serious AEs occurred in eight patients in both arms. Rescue medication was needed for 30 patients receiving sitagliptin and 11 patients receiving liraglutide. No cases of pancreatitis were reported. In the sitagliptin group, one subject each developed bladder cancer and squamous cell carcinoma. Nocturnal hypoglycemia did not develop in either trial arm.
Funding was provided by liraglutide maker Novo Nordisk. Dr. Maislos had no disclosures.
BOSTON – Switching from sitagliptin to liraglutide, in combination with metformin, improved control of hypoglycemia and resulted in greater weight loss in patients with type 2 diabetes, reported Dr. Maximo Maislos at the annual meeting of the Endocrine Society.
Results of a randomized, double-blind, double-dummy, active-controlled 26-week trial have indicated that liraglutide can be used as an add-on to metformin for patients with type 2 diabetes who have remained hyperglycemic.
“Switching from sitagliptin to liraglutide resulted in superior [glycated hemoglobin] and body weight reductions, compared with continued sitagliptin treatment,” said Dr. Maximo Maislos of Ben-Gurion University, Beer-Sheva, Israel.
The LIRA-SWITCH trial (Efficacy and Safety of Switching From Sitagliptin to Liraglutide in Subjects With Type 2 Diabetes Not Achieving Adequate Glycaemic Control on Sitagliptin and Metformin) involved 407 patients. The majority (60%) were male; mean age was 56 years and mean body mass index was 32 kg/m2. The subjects had all been treated with sitagliptin (100 mg/day) and metformin (greater than or equal to 1,500 mg/day or a maximum tolerated dose greater than or equal to 1,000 mg/day) for at least 90 days. Hyperglycemia had not been well controlled, with a mean hemoglobin A1C (HbAIC) level of 8.3% (67 mmol/mol). The mean duration of type 2 diabetes was 8 years.
Subjects were randomized to continued sitagliptin along with metformin (n = 204) or liraglutide (1.8 mg daily) along with metformin (n = 203).
After 26 weeks of treatment, reduction in HbAIC was significantly greater in the liraglutide arm than in the sitagliptin arm (1.14% vs. 0.54%; estimated treatment difference [ETD], –0.61%; 95% confidence interval, –0.82 to –0.40; P less than .0001). Those receiving liraglutide had statistically significantly greater weight loss, compared with those who continued on sitagliptin.
The less than 7% and less than or equal to 6.5% target levels of HbAIC were achieved by 50.6% and 29.5%, respectively, of patients in the liraglutide arm. These percentages were significantly higher than the respective 26.9% and 9.9% of patients in the sitagliptin arm (P less than .0001 for both). Fasting plasma glucose levels were significantly reduced with liraglutide treatment while decreases in systolic and diastolic blood pressure were similar in the two study arms.
Adverse events (AEs) occurred more often in the liraglutide group than in the sitagliptin group (68.8% vs. 56.9%). Thirteen patients receiving liraglutide discontinued treatment, compared with five in the sitagliptin arm. The most common AEs in the liraglutide group were gastrointestinal disorders, principally nausea (21.8% with liraglutide vs. 7.8% with sitagliptin) and diarrhea (16.3% with liraglutide vs. 9.3% with sitagliptin), followed by decreased appetite (8.9% vs. 3.4%, respectively). These AEs tended to subside within the first few weeks of treatment.
Serious AEs occurred in eight patients in both arms. Rescue medication was needed for 30 patients receiving sitagliptin and 11 patients receiving liraglutide. No cases of pancreatitis were reported. In the sitagliptin group, one subject each developed bladder cancer and squamous cell carcinoma. Nocturnal hypoglycemia did not develop in either trial arm.
Funding was provided by liraglutide maker Novo Nordisk. Dr. Maislos had no disclosures.
BOSTON – Switching from sitagliptin to liraglutide, in combination with metformin, improved control of hypoglycemia and resulted in greater weight loss in patients with type 2 diabetes, reported Dr. Maximo Maislos at the annual meeting of the Endocrine Society.
Results of a randomized, double-blind, double-dummy, active-controlled 26-week trial have indicated that liraglutide can be used as an add-on to metformin for patients with type 2 diabetes who have remained hyperglycemic.
“Switching from sitagliptin to liraglutide resulted in superior [glycated hemoglobin] and body weight reductions, compared with continued sitagliptin treatment,” said Dr. Maximo Maislos of Ben-Gurion University, Beer-Sheva, Israel.
The LIRA-SWITCH trial (Efficacy and Safety of Switching From Sitagliptin to Liraglutide in Subjects With Type 2 Diabetes Not Achieving Adequate Glycaemic Control on Sitagliptin and Metformin) involved 407 patients. The majority (60%) were male; mean age was 56 years and mean body mass index was 32 kg/m2. The subjects had all been treated with sitagliptin (100 mg/day) and metformin (greater than or equal to 1,500 mg/day or a maximum tolerated dose greater than or equal to 1,000 mg/day) for at least 90 days. Hyperglycemia had not been well controlled, with a mean hemoglobin A1C (HbAIC) level of 8.3% (67 mmol/mol). The mean duration of type 2 diabetes was 8 years.
Subjects were randomized to continued sitagliptin along with metformin (n = 204) or liraglutide (1.8 mg daily) along with metformin (n = 203).
After 26 weeks of treatment, reduction in HbAIC was significantly greater in the liraglutide arm than in the sitagliptin arm (1.14% vs. 0.54%; estimated treatment difference [ETD], –0.61%; 95% confidence interval, –0.82 to –0.40; P less than .0001). Those receiving liraglutide had statistically significantly greater weight loss, compared with those who continued on sitagliptin.
The less than 7% and less than or equal to 6.5% target levels of HbAIC were achieved by 50.6% and 29.5%, respectively, of patients in the liraglutide arm. These percentages were significantly higher than the respective 26.9% and 9.9% of patients in the sitagliptin arm (P less than .0001 for both). Fasting plasma glucose levels were significantly reduced with liraglutide treatment while decreases in systolic and diastolic blood pressure were similar in the two study arms.
Adverse events (AEs) occurred more often in the liraglutide group than in the sitagliptin group (68.8% vs. 56.9%). Thirteen patients receiving liraglutide discontinued treatment, compared with five in the sitagliptin arm. The most common AEs in the liraglutide group were gastrointestinal disorders, principally nausea (21.8% with liraglutide vs. 7.8% with sitagliptin) and diarrhea (16.3% with liraglutide vs. 9.3% with sitagliptin), followed by decreased appetite (8.9% vs. 3.4%, respectively). These AEs tended to subside within the first few weeks of treatment.
Serious AEs occurred in eight patients in both arms. Rescue medication was needed for 30 patients receiving sitagliptin and 11 patients receiving liraglutide. No cases of pancreatitis were reported. In the sitagliptin group, one subject each developed bladder cancer and squamous cell carcinoma. Nocturnal hypoglycemia did not develop in either trial arm.
Funding was provided by liraglutide maker Novo Nordisk. Dr. Maislos had no disclosures.
AT ENDO 2016
Key clinical point: A switch from sitagliptin to liraglutide provides an option, in combination with metformin, to improve the management of type 2 diabetes, allowing patients to remain on dual therapy.
Major finding: After 26 weeks of treatment, reduction in HbAIC was significantly greater in the liraglutide arm than in the sitagliptin arm (–1.14% vs. –0.54%. Those receiving liraglutide had statistically significantly greater weight loss, compared with those who continued on sitagliptin.
Data source: 407 patients with type 2 diabetes enrolled in a randomized, double-blind, double-dummy, active-controlled 26-week trial.
Disclosures: Dr. Maislos had no disclosures.