Nonpruritic rash on arms

Article Type
Changed
Fri, 01/18/2019 - 08:49
Display Headline
Nonpruritic rash on arms

Based on the results of the punch biopsy and the slight scale seen on the periphery of the lesions (a collarette scale pattern), the FP made a diagnosis of pityriasis rosea.

The distribution of the lesions in this case was not typical for pityriasis rosea; lesions are typically found on the trunk (not the arms) and may start with a herald patch. Given the distribution of the lesions in this case, the more precise diagnosis was inverse pityriasis rosea.

The physician explained to the patient and her mother that the rash would resolve spontaneously and was unlikely to leave any scarring. Six months later, the FP saw the mother for an unrelated issue and she said her daughter’s rash had gotten better within a month of her daughter’s visit, and there had been no scarring.

 

Photos and text for Photo Rounds Friday courtesy of Richard P. Usatine, MD. This case was adapted from: Henderson D, Usatine R. Pityriasis rosea. In: Usatine R, Smith M, Mayeaux EJ, et al, eds. Color Atlas of Family Medicine. 2nd ed. New York, NY: McGraw-Hill; 2013: 896-900.

To learn more about the Color Atlas of Family Medicine, see: www.amazon.com/Color-Family-Medicine-Richard-Usatine/dp/0071769641/

You can now get the second edition of the Color Atlas of Family Medicine as an app by clicking on this link: usatinemedia.com

Issue
The Journal of Family Practice - 66(9)
Publications
Topics
Sections

Based on the results of the punch biopsy and the slight scale seen on the periphery of the lesions (a collarette scale pattern), the FP made a diagnosis of pityriasis rosea.

The distribution of the lesions in this case was not typical for pityriasis rosea; lesions are typically found on the trunk (not the arms) and may start with a herald patch. Given the distribution of the lesions in this case, the more precise diagnosis was inverse pityriasis rosea.

The physician explained to the patient and her mother that the rash would resolve spontaneously and was unlikely to leave any scarring. Six months later, the FP saw the mother for an unrelated issue and she said her daughter’s rash had gotten better within a month of her daughter’s visit, and there had been no scarring.

 

Photos and text for Photo Rounds Friday courtesy of Richard P. Usatine, MD. This case was adapted from: Henderson D, Usatine R. Pityriasis rosea. In: Usatine R, Smith M, Mayeaux EJ, et al, eds. Color Atlas of Family Medicine. 2nd ed. New York, NY: McGraw-Hill; 2013: 896-900.

To learn more about the Color Atlas of Family Medicine, see: www.amazon.com/Color-Family-Medicine-Richard-Usatine/dp/0071769641/

You can now get the second edition of the Color Atlas of Family Medicine as an app by clicking on this link: usatinemedia.com

Based on the results of the punch biopsy and the slight scale seen on the periphery of the lesions (a collarette scale pattern), the FP made a diagnosis of pityriasis rosea.

The distribution of the lesions in this case was not typical for pityriasis rosea; lesions are typically found on the trunk (not the arms) and may start with a herald patch. Given the distribution of the lesions in this case, the more precise diagnosis was inverse pityriasis rosea.

The physician explained to the patient and her mother that the rash would resolve spontaneously and was unlikely to leave any scarring. Six months later, the FP saw the mother for an unrelated issue and she said her daughter’s rash had gotten better within a month of her daughter’s visit, and there had been no scarring.

 

Photos and text for Photo Rounds Friday courtesy of Richard P. Usatine, MD. This case was adapted from: Henderson D, Usatine R. Pityriasis rosea. In: Usatine R, Smith M, Mayeaux EJ, et al, eds. Color Atlas of Family Medicine. 2nd ed. New York, NY: McGraw-Hill; 2013: 896-900.

To learn more about the Color Atlas of Family Medicine, see: www.amazon.com/Color-Family-Medicine-Richard-Usatine/dp/0071769641/

You can now get the second edition of the Color Atlas of Family Medicine as an app by clicking on this link: usatinemedia.com

Issue
The Journal of Family Practice - 66(9)
Issue
The Journal of Family Practice - 66(9)
Publications
Publications
Topics
Article Type
Display Headline
Nonpruritic rash on arms
Display Headline
Nonpruritic rash on arms
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default

Delayed-release metformin proves promising for diabetic renal disease

Article Type
Changed
Tue, 05/03/2022 - 15:22

A delayed-release (DR) formulation of metformin produced improvements in glycated hemoglobin and fasting plasma glucose levels in a 16-week, dose-ranging, phase 2 trial performed in patients with type 2 diabetes mellitus and chronic kidney disease (CKD).

There was also a reduced incidence of gastrointestinal side effects with metformin DR versus immediate-release (IR) metformin (less than 16% at all doses tested vs. 28%), particularly with regard to nausea (1%-3% vs. 10%).

Sara Freeman/Frontline Medical News
Dr. Juan Pablo Frias
“Currently available metformin formulations, whether it be immediate release (IR) or extended release (ER), are almost exclusively eliminated by the kidneys,” said study investigator Juan Pablo Frias, MD, president and CEO of the National Research Institute, a privately owned multispecialty research facility in Los Angeles, at the annual meeting of the European Association for the Study of Diabetes.

“Based on concerns about the potential for lactic acidosis, primarily in patients with fairly severe renal impairment, metformin use is contraindicated in patients with stage IV chronic kidney disease,” Dr. Frias observed. Furthermore its use is restricted in patients with stage IIIB CKD, with European guidelines recommending lower starting (500 mg) and maximum (1,000 mg) daily doses, and U.S. guidelines recommending continuing treatment with caution in those already on metformin and not initiating metformin in new patients with this stage of kidney disease.

Treating patients with type 2 diabetes mellitus (T2DM) and later stages of CKD is challenging, as there are issues with almost all of the available alternatives to metformin, he noted. For instance, insulin use and sulfonylureas carry the risk of hypoglycemia, which is higher in patients with stage IIIB/IV renal disease than without. The dipeptidyl peptidase-4 inhibitors are “modestly able to reduce A1c, but generally do much better in combination with metformin, which is often contraindicated in these patients,” Dr. Frias said. Sodium glucose cotransporter 2 inhibitors are “generally not effective” in this patient group, he said.

Metformin DR is being specifically developed to manage patients with T2DM patients and stage IIIB/IV CKD, Dr. Frias said. Its enteric coating helps it bypass the stomach and upper intestine and so ensures that the majority of metformin absorption occurs in the lower bowel to reduce systemic exposure while retaining its positive effects on glycemic mechanisms such as the secretion of glucagon-like peptide 1.

In the current phase 2 study, 571 patients with T2DM and stage I/II CKD were recruited. Patients with stage IIIB/IV were not included because of the restrictions on the use of metformin.

Patients were randomized to receive placebo or metformin DR (600 mg, 900 mg, 1,200 mg, and 1,500 mg twice daily) in a double-blind comparison, with a single-blind reference arm of metformin IR, (1,000 mg once daily for the first week then 1,000 mg twice daily) also included as part of the study design.

The change in hemoglobin A1c (HbA1c) from baseline levels to week 16 of treatment, the primary endpoint, was significantly (P less than .05) greater with metformin DR) than with placebo (–0.49%, –0.62%, and –0.06%, respectively). Changes in fasting plasma glucose (FPG) from week 4 to week 16 were also higher with metformin DR than with placebo, with the 1,200 metformin DR dose achieving a 25.1 mg/dL drop in FPG, “almost 80% of the fasting glucose–lowering capacity of the immediate release formulation.”

While the changes in HbA1c (-1.10%) and FPG (-32.6 m/dL) were greatest with metformin IR, the lower systemic exposure needs to be considered, Dr. Frias said. The plasma exposure with metformin DR was less than 37% that of metformin IR.

“If we normalize for systemic exposure, so for any given unit, if you will, of systemic exposure, you actually had [a 1.5-fold] improved hemoglobin A1c with the delayed-release formulation, and a twofold increase in the fasting glucose,” Dr. Frias reported. “So from a practical point of view, if you needed to reach those ‘safe’ plasma concentrations with an immediate-release formulation, you would have to lower [the dose of] that formulation, probably to a dose that would not be efficacious for a patient.”

As for safety, any adverse event (AE) occurred in 41.7% of placebo-treated patients, in 47.9% of metformin IR–treated patients, and in 55.3%, 48.4%, 39.6%, and 43.8%, of those taking metformin DR at the respective doses of 600 mg, 900 mg, 1,200 mg, and 1, 500 mg.

Serious AEs were recorded in 4.2% of placebo-treated patients, 1.1% of metformin IR-treated patients, and in 1.1%, 0%, 4.2%, and 1.0% those taking increasing doses of metformin DR.

There were fewer AEs related to study medication (12.8%, 13.7%, 14.6%, and 9.4%) and subsequently resulting in discontinuation (3.2%, 2.1%, 7.3%, 2.1%) with metformin DR than with metformin IR (25.5%, 8.5%). Of placebo-treated patients, 6.3% developed a treatment-related AE, and 6.3% discontinued the study as a result.

“The improved risk/benefit profile that’s seen [in this study] would lead you to think that this would be a formulation that would be effective, particularly in patients with CKD IIIB or IV,” Dr. Frias concluded, noting that further studies would need to look into this possibility further.

The study was funded by Elcelyx Therapeutics. Dr. Frias disclosed receiving research support from Abbvie, Eli Lilly, IONIS, Janssen, Johnson & Johnson, Merck, Mylan, Novartis, Pfizer, and vTv therapeutics. He has also received research support from and participated in scientific advisory boards for AstraZeneca, Boehringer Ingelheim, Bristol-Myers Squibb, Novo Nordisk, and Theracos. A coauthor is an employee of Elcelyx Therapeutics and disclosed being a shareholder of the company.

 

 

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

A delayed-release (DR) formulation of metformin produced improvements in glycated hemoglobin and fasting plasma glucose levels in a 16-week, dose-ranging, phase 2 trial performed in patients with type 2 diabetes mellitus and chronic kidney disease (CKD).

There was also a reduced incidence of gastrointestinal side effects with metformin DR versus immediate-release (IR) metformin (less than 16% at all doses tested vs. 28%), particularly with regard to nausea (1%-3% vs. 10%).

Sara Freeman/Frontline Medical News
Dr. Juan Pablo Frias
“Currently available metformin formulations, whether it be immediate release (IR) or extended release (ER), are almost exclusively eliminated by the kidneys,” said study investigator Juan Pablo Frias, MD, president and CEO of the National Research Institute, a privately owned multispecialty research facility in Los Angeles, at the annual meeting of the European Association for the Study of Diabetes.

“Based on concerns about the potential for lactic acidosis, primarily in patients with fairly severe renal impairment, metformin use is contraindicated in patients with stage IV chronic kidney disease,” Dr. Frias observed. Furthermore its use is restricted in patients with stage IIIB CKD, with European guidelines recommending lower starting (500 mg) and maximum (1,000 mg) daily doses, and U.S. guidelines recommending continuing treatment with caution in those already on metformin and not initiating metformin in new patients with this stage of kidney disease.

Treating patients with type 2 diabetes mellitus (T2DM) and later stages of CKD is challenging, as there are issues with almost all of the available alternatives to metformin, he noted. For instance, insulin use and sulfonylureas carry the risk of hypoglycemia, which is higher in patients with stage IIIB/IV renal disease than without. The dipeptidyl peptidase-4 inhibitors are “modestly able to reduce A1c, but generally do much better in combination with metformin, which is often contraindicated in these patients,” Dr. Frias said. Sodium glucose cotransporter 2 inhibitors are “generally not effective” in this patient group, he said.

Metformin DR is being specifically developed to manage patients with T2DM patients and stage IIIB/IV CKD, Dr. Frias said. Its enteric coating helps it bypass the stomach and upper intestine and so ensures that the majority of metformin absorption occurs in the lower bowel to reduce systemic exposure while retaining its positive effects on glycemic mechanisms such as the secretion of glucagon-like peptide 1.

In the current phase 2 study, 571 patients with T2DM and stage I/II CKD were recruited. Patients with stage IIIB/IV were not included because of the restrictions on the use of metformin.

Patients were randomized to receive placebo or metformin DR (600 mg, 900 mg, 1,200 mg, and 1,500 mg twice daily) in a double-blind comparison, with a single-blind reference arm of metformin IR, (1,000 mg once daily for the first week then 1,000 mg twice daily) also included as part of the study design.

The change in hemoglobin A1c (HbA1c) from baseline levels to week 16 of treatment, the primary endpoint, was significantly (P less than .05) greater with metformin DR) than with placebo (–0.49%, –0.62%, and –0.06%, respectively). Changes in fasting plasma glucose (FPG) from week 4 to week 16 were also higher with metformin DR than with placebo, with the 1,200 metformin DR dose achieving a 25.1 mg/dL drop in FPG, “almost 80% of the fasting glucose–lowering capacity of the immediate release formulation.”

While the changes in HbA1c (-1.10%) and FPG (-32.6 m/dL) were greatest with metformin IR, the lower systemic exposure needs to be considered, Dr. Frias said. The plasma exposure with metformin DR was less than 37% that of metformin IR.

“If we normalize for systemic exposure, so for any given unit, if you will, of systemic exposure, you actually had [a 1.5-fold] improved hemoglobin A1c with the delayed-release formulation, and a twofold increase in the fasting glucose,” Dr. Frias reported. “So from a practical point of view, if you needed to reach those ‘safe’ plasma concentrations with an immediate-release formulation, you would have to lower [the dose of] that formulation, probably to a dose that would not be efficacious for a patient.”

As for safety, any adverse event (AE) occurred in 41.7% of placebo-treated patients, in 47.9% of metformin IR–treated patients, and in 55.3%, 48.4%, 39.6%, and 43.8%, of those taking metformin DR at the respective doses of 600 mg, 900 mg, 1,200 mg, and 1, 500 mg.

Serious AEs were recorded in 4.2% of placebo-treated patients, 1.1% of metformin IR-treated patients, and in 1.1%, 0%, 4.2%, and 1.0% those taking increasing doses of metformin DR.

There were fewer AEs related to study medication (12.8%, 13.7%, 14.6%, and 9.4%) and subsequently resulting in discontinuation (3.2%, 2.1%, 7.3%, 2.1%) with metformin DR than with metformin IR (25.5%, 8.5%). Of placebo-treated patients, 6.3% developed a treatment-related AE, and 6.3% discontinued the study as a result.

“The improved risk/benefit profile that’s seen [in this study] would lead you to think that this would be a formulation that would be effective, particularly in patients with CKD IIIB or IV,” Dr. Frias concluded, noting that further studies would need to look into this possibility further.

The study was funded by Elcelyx Therapeutics. Dr. Frias disclosed receiving research support from Abbvie, Eli Lilly, IONIS, Janssen, Johnson & Johnson, Merck, Mylan, Novartis, Pfizer, and vTv therapeutics. He has also received research support from and participated in scientific advisory boards for AstraZeneca, Boehringer Ingelheim, Bristol-Myers Squibb, Novo Nordisk, and Theracos. A coauthor is an employee of Elcelyx Therapeutics and disclosed being a shareholder of the company.

 

 

A delayed-release (DR) formulation of metformin produced improvements in glycated hemoglobin and fasting plasma glucose levels in a 16-week, dose-ranging, phase 2 trial performed in patients with type 2 diabetes mellitus and chronic kidney disease (CKD).

There was also a reduced incidence of gastrointestinal side effects with metformin DR versus immediate-release (IR) metformin (less than 16% at all doses tested vs. 28%), particularly with regard to nausea (1%-3% vs. 10%).

Sara Freeman/Frontline Medical News
Dr. Juan Pablo Frias
“Currently available metformin formulations, whether it be immediate release (IR) or extended release (ER), are almost exclusively eliminated by the kidneys,” said study investigator Juan Pablo Frias, MD, president and CEO of the National Research Institute, a privately owned multispecialty research facility in Los Angeles, at the annual meeting of the European Association for the Study of Diabetes.

“Based on concerns about the potential for lactic acidosis, primarily in patients with fairly severe renal impairment, metformin use is contraindicated in patients with stage IV chronic kidney disease,” Dr. Frias observed. Furthermore its use is restricted in patients with stage IIIB CKD, with European guidelines recommending lower starting (500 mg) and maximum (1,000 mg) daily doses, and U.S. guidelines recommending continuing treatment with caution in those already on metformin and not initiating metformin in new patients with this stage of kidney disease.

Treating patients with type 2 diabetes mellitus (T2DM) and later stages of CKD is challenging, as there are issues with almost all of the available alternatives to metformin, he noted. For instance, insulin use and sulfonylureas carry the risk of hypoglycemia, which is higher in patients with stage IIIB/IV renal disease than without. The dipeptidyl peptidase-4 inhibitors are “modestly able to reduce A1c, but generally do much better in combination with metformin, which is often contraindicated in these patients,” Dr. Frias said. Sodium glucose cotransporter 2 inhibitors are “generally not effective” in this patient group, he said.

Metformin DR is being specifically developed to manage patients with T2DM patients and stage IIIB/IV CKD, Dr. Frias said. Its enteric coating helps it bypass the stomach and upper intestine and so ensures that the majority of metformin absorption occurs in the lower bowel to reduce systemic exposure while retaining its positive effects on glycemic mechanisms such as the secretion of glucagon-like peptide 1.

In the current phase 2 study, 571 patients with T2DM and stage I/II CKD were recruited. Patients with stage IIIB/IV were not included because of the restrictions on the use of metformin.

Patients were randomized to receive placebo or metformin DR (600 mg, 900 mg, 1,200 mg, and 1,500 mg twice daily) in a double-blind comparison, with a single-blind reference arm of metformin IR, (1,000 mg once daily for the first week then 1,000 mg twice daily) also included as part of the study design.

The change in hemoglobin A1c (HbA1c) from baseline levels to week 16 of treatment, the primary endpoint, was significantly (P less than .05) greater with metformin DR) than with placebo (–0.49%, –0.62%, and –0.06%, respectively). Changes in fasting plasma glucose (FPG) from week 4 to week 16 were also higher with metformin DR than with placebo, with the 1,200 metformin DR dose achieving a 25.1 mg/dL drop in FPG, “almost 80% of the fasting glucose–lowering capacity of the immediate release formulation.”

While the changes in HbA1c (-1.10%) and FPG (-32.6 m/dL) were greatest with metformin IR, the lower systemic exposure needs to be considered, Dr. Frias said. The plasma exposure with metformin DR was less than 37% that of metformin IR.

“If we normalize for systemic exposure, so for any given unit, if you will, of systemic exposure, you actually had [a 1.5-fold] improved hemoglobin A1c with the delayed-release formulation, and a twofold increase in the fasting glucose,” Dr. Frias reported. “So from a practical point of view, if you needed to reach those ‘safe’ plasma concentrations with an immediate-release formulation, you would have to lower [the dose of] that formulation, probably to a dose that would not be efficacious for a patient.”

As for safety, any adverse event (AE) occurred in 41.7% of placebo-treated patients, in 47.9% of metformin IR–treated patients, and in 55.3%, 48.4%, 39.6%, and 43.8%, of those taking metformin DR at the respective doses of 600 mg, 900 mg, 1,200 mg, and 1, 500 mg.

Serious AEs were recorded in 4.2% of placebo-treated patients, 1.1% of metformin IR-treated patients, and in 1.1%, 0%, 4.2%, and 1.0% those taking increasing doses of metformin DR.

There were fewer AEs related to study medication (12.8%, 13.7%, 14.6%, and 9.4%) and subsequently resulting in discontinuation (3.2%, 2.1%, 7.3%, 2.1%) with metformin DR than with metformin IR (25.5%, 8.5%). Of placebo-treated patients, 6.3% developed a treatment-related AE, and 6.3% discontinued the study as a result.

“The improved risk/benefit profile that’s seen [in this study] would lead you to think that this would be a formulation that would be effective, particularly in patients with CKD IIIB or IV,” Dr. Frias concluded, noting that further studies would need to look into this possibility further.

The study was funded by Elcelyx Therapeutics. Dr. Frias disclosed receiving research support from Abbvie, Eli Lilly, IONIS, Janssen, Johnson & Johnson, Merck, Mylan, Novartis, Pfizer, and vTv therapeutics. He has also received research support from and participated in scientific advisory boards for AstraZeneca, Boehringer Ingelheim, Bristol-Myers Squibb, Novo Nordisk, and Theracos. A coauthor is an employee of Elcelyx Therapeutics and disclosed being a shareholder of the company.

 

 

Publications
Publications
Topics
Article Type
Sections
Article Source

AT EASD 2017

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

 

Key clinical point: A delayed-release formulation of metformin appears have a risk/benefit profile that would enable use in patients with type 2 diabetes and chronic kidney disease.

Major finding: Change in HbA1c at week 16 (primary endpoint) was –0.49%, –0.62%, and –0.06%, for metformin DR 1,200 mg, 1,500 mg, and placebo, respectively, P less than .05).

Data source: A 16-week, dose-ranging phase 2 trial involving 571 patients with T2DM and CKD.

Disclosures: The study was funded by Elcelyx Therapeutics. Dr. Frias disclosed receiving research support from Abbvie, Eli Lilly, IONIS, Janssen, Johnson & Johnson, Merck, Mylan, Novartis, Pfizer, and vTv therapeutics. He has also received research support from and participated in scientific advisory boards for AstraZeneca, Boehringer Ingelheim, Bristol-Myers Squibb, Novo Nordisk, and Theracos. A coauthor is an employee of Elcelyx Therapeutics and disclosed being a shareholder of the company.

Disqus Comments
Default

Rituximab maintenance halves MCL death risk after ASCT

Article Type
Changed
Fri, 12/16/2022 - 12:37

 

Patients with mantle cell lymphoma who received maintenance therapy with rituximab following autologous stem cell transplantation (ASCT) had significantly better overall survival, event-free survival, and progression-free survival, compared with patients who were followed with observation alone after transplant, results of a phase 3 trial show.

After 50.2 months median follow-up, the overall survival rate for patients aged 65 or younger randomized to rituximab maintenance after four cycles of induction chemotherapy with rituximab, dexamethasone, cytarabine, and a platinum derivative (R-DHAP) followed by ASCT was 89%, compared with 80% for patients randomized to observation (P = .004), reported Steven Le Gouill, MD, PhD, of University Hospital Hotel-Dieu, in Nantes, Frances, and colleagues.

In an unadjusted regression analysis, the difference translated into a hazard ratio for death within 4 years of 0.50 (P = .004) favoring rituximab, they wrote in the Sept. 28, 2017 issue of The New England Journal of Medicine.

“[A]n induction regimen with four courses of R-DHAP followed by transplantation without total-body irradiation resulted in a high rate of complete response. A 3-year course of rituximab maintenance therapy administered every 2 months prolonged overall survival among young patients with mantle cell lymphoma,” the investigators wrote (N Engl J Med. 2017;377:1250-60).

Dr. Le Gouill and his colleagues hypothesized that relapses following treatment for MCL may be caused by residual malignant cells that chemotherapy and ASCT fail to eradicate, suggesting that maintenance therapy with rituximab could help to suppress residual disease, prolong the duration of responses, and extend both progression-free and overall survival.

They cited an earlier study by members of the European Mantle Cell Lymphoma Network showing that among patients aged 60 and older who had a response to eight cycles of chemotherapy with rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP), maintenance therapy with rituximab was associated with an 87% 4-year overall survival rate vs. 63% for patients maintained on interferon alfa (P = .005) (N Engl J Med. 2012;367:520-31).

For the current study, the researchers enrolled 299 patients, of whom 257 went on to ASCT, and 240 of whom were randomized and were included in an intention-to-treat (ITT) analysis.

Patients received induction with four cycles of R-DHAP. Those patients who had partial responses or tumor mass shrinkage of less than 75% on CT received a rescue induction with four cycles of R-CHOP.

Those patients with complete or partial responses could then go on to transplantation after a conditioning regimen of R-BEAM (rituximab, carmustine, etoposide, cytarabine, and melphalan).

Patients randomized after ASCT to rituximab received it every 2 months for 3 years in an intravenous infusion at a dose of 375 mg/m2.

After a median of 50.2 months from randomization, the rate of 4-year event-free survival (no disease progression, relapse, death, or severe infection), the primary endpoint, was 79% for patients maintained on rituximab vs. 61% for those on observation alone (P = .001).

The 4-year progression-free survival rate also favored rituximab at 83% vs. 64%, respectively (P less than .001), with respective overall survival rates of 89% and 80%.

The median event-free survival, progression-free survival, and overall survival was not reached in either study arm.

For the 59 patients who for various reasons did not undergo randomization, the median progression-free survival was 11.0 months, and the median overall survival was 30.6 months.

In all, 83 of the 120 patients randomized to rituximab completed the scheduled 3 years of therapy. Maintenance therapy was stopped for disease progression in 16 patients and because of neutropenia in 9. There were 13 deaths in the rituximab arm, including 3 deaths from second malignancies.

Of the 120 patients assigned to observation, 37 had disease progression during the study period, and 24 died, one from a second malignancy.

Four patients in each study arm had serious infections after ASCT, including one case each of spondylitis, pyelonephritis, septicemia, and varicella pneumonia in the rituximab group, and septicemia, cellulitis, meningitis, and severe pneumonia in the observation group.

Lymphoma was the cause of death in 8 patients assigned to rituximab, and in 16 assigned to observation.

The investigators noted that although some centers use total-body irradiation for conditioning prior to transplant, this modality is not available in all centers and is associated with both short- and long-term toxicities. The progression-free survival results seen in this trial, where only ablative drug regimens were used “suggest that total-body irradiation–based conditioning regimens may not be superior to chemotherapy alone when an effective regimen is used during induction,” they wrote.

The study was supported by Roche and Amgen. Dr. Le Gouill disclosed fees for consulting and honoraria from Roche, Janssen-Cilag, and Celgene. Multiple coauthors disclosed similar relationships with industry.

Publications
Topics
Sections

 

Patients with mantle cell lymphoma who received maintenance therapy with rituximab following autologous stem cell transplantation (ASCT) had significantly better overall survival, event-free survival, and progression-free survival, compared with patients who were followed with observation alone after transplant, results of a phase 3 trial show.

After 50.2 months median follow-up, the overall survival rate for patients aged 65 or younger randomized to rituximab maintenance after four cycles of induction chemotherapy with rituximab, dexamethasone, cytarabine, and a platinum derivative (R-DHAP) followed by ASCT was 89%, compared with 80% for patients randomized to observation (P = .004), reported Steven Le Gouill, MD, PhD, of University Hospital Hotel-Dieu, in Nantes, Frances, and colleagues.

In an unadjusted regression analysis, the difference translated into a hazard ratio for death within 4 years of 0.50 (P = .004) favoring rituximab, they wrote in the Sept. 28, 2017 issue of The New England Journal of Medicine.

“[A]n induction regimen with four courses of R-DHAP followed by transplantation without total-body irradiation resulted in a high rate of complete response. A 3-year course of rituximab maintenance therapy administered every 2 months prolonged overall survival among young patients with mantle cell lymphoma,” the investigators wrote (N Engl J Med. 2017;377:1250-60).

Dr. Le Gouill and his colleagues hypothesized that relapses following treatment for MCL may be caused by residual malignant cells that chemotherapy and ASCT fail to eradicate, suggesting that maintenance therapy with rituximab could help to suppress residual disease, prolong the duration of responses, and extend both progression-free and overall survival.

They cited an earlier study by members of the European Mantle Cell Lymphoma Network showing that among patients aged 60 and older who had a response to eight cycles of chemotherapy with rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP), maintenance therapy with rituximab was associated with an 87% 4-year overall survival rate vs. 63% for patients maintained on interferon alfa (P = .005) (N Engl J Med. 2012;367:520-31).

For the current study, the researchers enrolled 299 patients, of whom 257 went on to ASCT, and 240 of whom were randomized and were included in an intention-to-treat (ITT) analysis.

Patients received induction with four cycles of R-DHAP. Those patients who had partial responses or tumor mass shrinkage of less than 75% on CT received a rescue induction with four cycles of R-CHOP.

Those patients with complete or partial responses could then go on to transplantation after a conditioning regimen of R-BEAM (rituximab, carmustine, etoposide, cytarabine, and melphalan).

Patients randomized after ASCT to rituximab received it every 2 months for 3 years in an intravenous infusion at a dose of 375 mg/m2.

After a median of 50.2 months from randomization, the rate of 4-year event-free survival (no disease progression, relapse, death, or severe infection), the primary endpoint, was 79% for patients maintained on rituximab vs. 61% for those on observation alone (P = .001).

The 4-year progression-free survival rate also favored rituximab at 83% vs. 64%, respectively (P less than .001), with respective overall survival rates of 89% and 80%.

The median event-free survival, progression-free survival, and overall survival was not reached in either study arm.

For the 59 patients who for various reasons did not undergo randomization, the median progression-free survival was 11.0 months, and the median overall survival was 30.6 months.

In all, 83 of the 120 patients randomized to rituximab completed the scheduled 3 years of therapy. Maintenance therapy was stopped for disease progression in 16 patients and because of neutropenia in 9. There were 13 deaths in the rituximab arm, including 3 deaths from second malignancies.

Of the 120 patients assigned to observation, 37 had disease progression during the study period, and 24 died, one from a second malignancy.

Four patients in each study arm had serious infections after ASCT, including one case each of spondylitis, pyelonephritis, septicemia, and varicella pneumonia in the rituximab group, and septicemia, cellulitis, meningitis, and severe pneumonia in the observation group.

Lymphoma was the cause of death in 8 patients assigned to rituximab, and in 16 assigned to observation.

The investigators noted that although some centers use total-body irradiation for conditioning prior to transplant, this modality is not available in all centers and is associated with both short- and long-term toxicities. The progression-free survival results seen in this trial, where only ablative drug regimens were used “suggest that total-body irradiation–based conditioning regimens may not be superior to chemotherapy alone when an effective regimen is used during induction,” they wrote.

The study was supported by Roche and Amgen. Dr. Le Gouill disclosed fees for consulting and honoraria from Roche, Janssen-Cilag, and Celgene. Multiple coauthors disclosed similar relationships with industry.

 

Patients with mantle cell lymphoma who received maintenance therapy with rituximab following autologous stem cell transplantation (ASCT) had significantly better overall survival, event-free survival, and progression-free survival, compared with patients who were followed with observation alone after transplant, results of a phase 3 trial show.

After 50.2 months median follow-up, the overall survival rate for patients aged 65 or younger randomized to rituximab maintenance after four cycles of induction chemotherapy with rituximab, dexamethasone, cytarabine, and a platinum derivative (R-DHAP) followed by ASCT was 89%, compared with 80% for patients randomized to observation (P = .004), reported Steven Le Gouill, MD, PhD, of University Hospital Hotel-Dieu, in Nantes, Frances, and colleagues.

In an unadjusted regression analysis, the difference translated into a hazard ratio for death within 4 years of 0.50 (P = .004) favoring rituximab, they wrote in the Sept. 28, 2017 issue of The New England Journal of Medicine.

“[A]n induction regimen with four courses of R-DHAP followed by transplantation without total-body irradiation resulted in a high rate of complete response. A 3-year course of rituximab maintenance therapy administered every 2 months prolonged overall survival among young patients with mantle cell lymphoma,” the investigators wrote (N Engl J Med. 2017;377:1250-60).

Dr. Le Gouill and his colleagues hypothesized that relapses following treatment for MCL may be caused by residual malignant cells that chemotherapy and ASCT fail to eradicate, suggesting that maintenance therapy with rituximab could help to suppress residual disease, prolong the duration of responses, and extend both progression-free and overall survival.

They cited an earlier study by members of the European Mantle Cell Lymphoma Network showing that among patients aged 60 and older who had a response to eight cycles of chemotherapy with rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP), maintenance therapy with rituximab was associated with an 87% 4-year overall survival rate vs. 63% for patients maintained on interferon alfa (P = .005) (N Engl J Med. 2012;367:520-31).

For the current study, the researchers enrolled 299 patients, of whom 257 went on to ASCT, and 240 of whom were randomized and were included in an intention-to-treat (ITT) analysis.

Patients received induction with four cycles of R-DHAP. Those patients who had partial responses or tumor mass shrinkage of less than 75% on CT received a rescue induction with four cycles of R-CHOP.

Those patients with complete or partial responses could then go on to transplantation after a conditioning regimen of R-BEAM (rituximab, carmustine, etoposide, cytarabine, and melphalan).

Patients randomized after ASCT to rituximab received it every 2 months for 3 years in an intravenous infusion at a dose of 375 mg/m2.

After a median of 50.2 months from randomization, the rate of 4-year event-free survival (no disease progression, relapse, death, or severe infection), the primary endpoint, was 79% for patients maintained on rituximab vs. 61% for those on observation alone (P = .001).

The 4-year progression-free survival rate also favored rituximab at 83% vs. 64%, respectively (P less than .001), with respective overall survival rates of 89% and 80%.

The median event-free survival, progression-free survival, and overall survival was not reached in either study arm.

For the 59 patients who for various reasons did not undergo randomization, the median progression-free survival was 11.0 months, and the median overall survival was 30.6 months.

In all, 83 of the 120 patients randomized to rituximab completed the scheduled 3 years of therapy. Maintenance therapy was stopped for disease progression in 16 patients and because of neutropenia in 9. There were 13 deaths in the rituximab arm, including 3 deaths from second malignancies.

Of the 120 patients assigned to observation, 37 had disease progression during the study period, and 24 died, one from a second malignancy.

Four patients in each study arm had serious infections after ASCT, including one case each of spondylitis, pyelonephritis, septicemia, and varicella pneumonia in the rituximab group, and septicemia, cellulitis, meningitis, and severe pneumonia in the observation group.

Lymphoma was the cause of death in 8 patients assigned to rituximab, and in 16 assigned to observation.

The investigators noted that although some centers use total-body irradiation for conditioning prior to transplant, this modality is not available in all centers and is associated with both short- and long-term toxicities. The progression-free survival results seen in this trial, where only ablative drug regimens were used “suggest that total-body irradiation–based conditioning regimens may not be superior to chemotherapy alone when an effective regimen is used during induction,” they wrote.

The study was supported by Roche and Amgen. Dr. Le Gouill disclosed fees for consulting and honoraria from Roche, Janssen-Cilag, and Celgene. Multiple coauthors disclosed similar relationships with industry.

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM NEW ENGLAND JOURNAL OF MEDICINE

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

 

Key clinical point: Following stem cell transplantation, rituximab maintenance cut in half the risk for death in patients with mantle cell lymphoma.

Major finding: Four-year overall survival was 89% with rituximab maintenance vs. 80% for observation alone.

Data source: Randomized phase 3 trial in 240 patients aged 65 and younger at diagnosis of mantle cell lymphoma.

Disclosures: The study was supported by Roche and Amgen. Dr. Le Gouill disclosed fees for consulting and honoraria from Roche, Janssen-Cilag, and Celgene. Multiple coauthors disclosed similar relationships with industry.

Disqus Comments
Default

Tips for Living With Ataxia

Article Type
Changed
Mon, 01/07/2019 - 10:34

Click here to download the PDF.

Article PDF
Issue
Neurology Reviews - 25(10)
Publications
Topics
Page Number
24
Sections
Article PDF
Article PDF

Click here to download the PDF.

Click here to download the PDF.

Issue
Neurology Reviews - 25(10)
Issue
Neurology Reviews - 25(10)
Page Number
24
Page Number
24
Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Article PDF Media

FDA approves first duodenoscope with disposable distal cap

Article Type
Changed
Wed, 09/27/2017 - 16:05

 

The Food and Drug Administration has approved the Pentax ED34-i10T model duodenoscope, the first with a disposable distal cap, according to an FDA press release.

A disposable distal cap will improve the ability to clean and reprocess the duodenoscope. Without being thoroughly cleaned and disinfected, contaminated tissue can remain and potentially can be transmitted to other patients.

Purple FDA logo.
“We believe the new disposable distal cap represents a major step toward lowering the risk of future infections associated with these devices,” William Maisel, MD, acting director of the Office of Device Evaluation at the FDA Center for Devices and Radiological Health, said in a statement. “Improving the safety of duodenoscopes is a top priority for the FDA, and we encourage companies to continue to pursue innovations that will help reduce the risk to patients.”

A previous version of the Pentax duodenoscope, the ED-3490TK, was subject to a January 2017 FDA Safety Alert, because of the potential for cracks and gaps to develop in the adhesive sealing the duodenoscope’s distal cap.

“Since the issue of duodenoscope-associated transmission of infections first received widespread attention in 2015, the AGA Center for GI Innovation and Technology has been working closely with regulators and endoscope manufacturers to identify and address problems in scope design and develop a path forward to ensure zero device-associated infections,” said V. Raman Muthusamy, MD, AGAF, FACG, FASGE, chair, AGA Center for GI Innovation and Technology. “We applaud Pentax for answering our call for innovation to improve patient safety for this common and life-saving GI procedure. We encourage all device manufacturers to continue on a path of innovation to better support gastroenterologists and, most importantly, the patients we serve.”

Publications
Topics
Sections

 

The Food and Drug Administration has approved the Pentax ED34-i10T model duodenoscope, the first with a disposable distal cap, according to an FDA press release.

A disposable distal cap will improve the ability to clean and reprocess the duodenoscope. Without being thoroughly cleaned and disinfected, contaminated tissue can remain and potentially can be transmitted to other patients.

Purple FDA logo.
“We believe the new disposable distal cap represents a major step toward lowering the risk of future infections associated with these devices,” William Maisel, MD, acting director of the Office of Device Evaluation at the FDA Center for Devices and Radiological Health, said in a statement. “Improving the safety of duodenoscopes is a top priority for the FDA, and we encourage companies to continue to pursue innovations that will help reduce the risk to patients.”

A previous version of the Pentax duodenoscope, the ED-3490TK, was subject to a January 2017 FDA Safety Alert, because of the potential for cracks and gaps to develop in the adhesive sealing the duodenoscope’s distal cap.

“Since the issue of duodenoscope-associated transmission of infections first received widespread attention in 2015, the AGA Center for GI Innovation and Technology has been working closely with regulators and endoscope manufacturers to identify and address problems in scope design and develop a path forward to ensure zero device-associated infections,” said V. Raman Muthusamy, MD, AGAF, FACG, FASGE, chair, AGA Center for GI Innovation and Technology. “We applaud Pentax for answering our call for innovation to improve patient safety for this common and life-saving GI procedure. We encourage all device manufacturers to continue on a path of innovation to better support gastroenterologists and, most importantly, the patients we serve.”

 

The Food and Drug Administration has approved the Pentax ED34-i10T model duodenoscope, the first with a disposable distal cap, according to an FDA press release.

A disposable distal cap will improve the ability to clean and reprocess the duodenoscope. Without being thoroughly cleaned and disinfected, contaminated tissue can remain and potentially can be transmitted to other patients.

Purple FDA logo.
“We believe the new disposable distal cap represents a major step toward lowering the risk of future infections associated with these devices,” William Maisel, MD, acting director of the Office of Device Evaluation at the FDA Center for Devices and Radiological Health, said in a statement. “Improving the safety of duodenoscopes is a top priority for the FDA, and we encourage companies to continue to pursue innovations that will help reduce the risk to patients.”

A previous version of the Pentax duodenoscope, the ED-3490TK, was subject to a January 2017 FDA Safety Alert, because of the potential for cracks and gaps to develop in the adhesive sealing the duodenoscope’s distal cap.

“Since the issue of duodenoscope-associated transmission of infections first received widespread attention in 2015, the AGA Center for GI Innovation and Technology has been working closely with regulators and endoscope manufacturers to identify and address problems in scope design and develop a path forward to ensure zero device-associated infections,” said V. Raman Muthusamy, MD, AGAF, FACG, FASGE, chair, AGA Center for GI Innovation and Technology. “We applaud Pentax for answering our call for innovation to improve patient safety for this common and life-saving GI procedure. We encourage all device manufacturers to continue on a path of innovation to better support gastroenterologists and, most importantly, the patients we serve.”

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

Caffeine offers no perks for Parkinson’s patients

Turn attention to other PD treatments
Article Type
Changed
Mon, 01/07/2019 - 13:00

 

Caffeine consumption has no significant impact on motor skills in patients with Parkinson’s disease, based on data from a double-blind, randomized, placebo-controlled trial of 121 adults. The findings were published online Sept. 27 in Neurology.

Dr. Ronald B. Postuma
In this study, 60 patients received 200 mg of caffeine in capsule form twice daily, in the morning and after lunch for 6 months; 61 received a placebo. The amount of caffeine was approximately equal to three cups of coffee.

Motor skills worsened in the caffeine group by an average of 0.16 points on the Movement Disorder Society–sponsored Unified Parkinson’s Disease Rating Scale (MDS-UPDRS) part III during patients’ “on” state and by 0.64 points in the placebo group; the difference was not significant.

The researchers found no differences between groups in the clinical global impression of change based on both patient and examiner assessment. In addition, no differences appeared in depression or anxiety, or in quality of life.

The study findings included long-term follow up data from 88 patients assessed at 12 months and 66 patients assessed at 18 months. The results were similar to the 6-month results, and the study was stopped, although the original design included a 4-year extension.

A total of 29 patients in the caffeine group and 31 patients in the placebo group reported adverse events, and 7 patients in the caffeine group and 5 in the placebo group discontinued the study because of side effects. A serious adverse event was reported by one patient in each group, but neither was deemed related to the interventions.

The findings contrast with the effects of caffeine in the 6-week study in 2012 that showed a significant, 3.2-point improvement in motor skills on the MDS-UPDRS part III with caffeine use, as well as reduced daytime sleepiness, the researchers said (Neurology. 2012;79:651-8). Interpretations of the different findings between the trials may be constrained by factors including differences in the study populations, speed of dose escalation, and trial duration and the possible short-term nature of caffeine’s impact, they noted. “Regardless, our core finding is that caffeine cannot be recommended as symptomatic therapy for parkinsonism.” However, “since caffeine is safe and generally well tolerated, it seems reasonable to empirically try intermittent moderate doses of caffeine for somnolence, and repeat if improvement is seen,” they added.

Dr. Postuma disclosed grant funding from the Canadian Institute of Health Research, the Webster Foundation, and Fonds de Recherche du Québec-Santé for this study.

Body

 

The appeal of an inexpensive, well-tolerated intervention such as caffeine to improve motor symptoms in Parkinson’s patients gave Dr. Postuma and his associates a good reason to try to replicate the positive results they found in an earlier study of caffeine.

The investigators found no benefit from caffeine, although their study was designed to have more than four times as many participants as the pilot study, was adequately powered to detect the same level of improvement as before, and was planned to have an extended follow-up period to look for persistent effects. The trial ended early and enrolled approximately half of its intended total, but was well run and did not suffer from differential compliance or loss to follow-up.

Although the small number of participants resulted in a wide confidence interval and cannot exclude a small effect, this trial suggests that caffeine does not significantly improve Parkinson’s disease symptoms and that it should not be a priority for future Parkinson’s disease intervention studies.

Charles B. Hall, PhD, is affiliated with the department of epidemiology and population health at Albert Einstein College of Medicine, New York. He disclosed salary support from the National Institute of Occupational Safety and Health, National Institute of Aging, National Cancer Institute, and National Center for Advancing Translational Sciences. His comments are derived from an editorial that accompanied Dr. Postuma and colleagues’ study (Neurology. 2017 Sep 27. doi: 10.1212/WNL.0000000000004584).

Publications
Topics
Sections
Body

 

The appeal of an inexpensive, well-tolerated intervention such as caffeine to improve motor symptoms in Parkinson’s patients gave Dr. Postuma and his associates a good reason to try to replicate the positive results they found in an earlier study of caffeine.

The investigators found no benefit from caffeine, although their study was designed to have more than four times as many participants as the pilot study, was adequately powered to detect the same level of improvement as before, and was planned to have an extended follow-up period to look for persistent effects. The trial ended early and enrolled approximately half of its intended total, but was well run and did not suffer from differential compliance or loss to follow-up.

Although the small number of participants resulted in a wide confidence interval and cannot exclude a small effect, this trial suggests that caffeine does not significantly improve Parkinson’s disease symptoms and that it should not be a priority for future Parkinson’s disease intervention studies.

Charles B. Hall, PhD, is affiliated with the department of epidemiology and population health at Albert Einstein College of Medicine, New York. He disclosed salary support from the National Institute of Occupational Safety and Health, National Institute of Aging, National Cancer Institute, and National Center for Advancing Translational Sciences. His comments are derived from an editorial that accompanied Dr. Postuma and colleagues’ study (Neurology. 2017 Sep 27. doi: 10.1212/WNL.0000000000004584).

Body

 

The appeal of an inexpensive, well-tolerated intervention such as caffeine to improve motor symptoms in Parkinson’s patients gave Dr. Postuma and his associates a good reason to try to replicate the positive results they found in an earlier study of caffeine.

The investigators found no benefit from caffeine, although their study was designed to have more than four times as many participants as the pilot study, was adequately powered to detect the same level of improvement as before, and was planned to have an extended follow-up period to look for persistent effects. The trial ended early and enrolled approximately half of its intended total, but was well run and did not suffer from differential compliance or loss to follow-up.

Although the small number of participants resulted in a wide confidence interval and cannot exclude a small effect, this trial suggests that caffeine does not significantly improve Parkinson’s disease symptoms and that it should not be a priority for future Parkinson’s disease intervention studies.

Charles B. Hall, PhD, is affiliated with the department of epidemiology and population health at Albert Einstein College of Medicine, New York. He disclosed salary support from the National Institute of Occupational Safety and Health, National Institute of Aging, National Cancer Institute, and National Center for Advancing Translational Sciences. His comments are derived from an editorial that accompanied Dr. Postuma and colleagues’ study (Neurology. 2017 Sep 27. doi: 10.1212/WNL.0000000000004584).

Title
Turn attention to other PD treatments
Turn attention to other PD treatments

 

Caffeine consumption has no significant impact on motor skills in patients with Parkinson’s disease, based on data from a double-blind, randomized, placebo-controlled trial of 121 adults. The findings were published online Sept. 27 in Neurology.

Dr. Ronald B. Postuma
In this study, 60 patients received 200 mg of caffeine in capsule form twice daily, in the morning and after lunch for 6 months; 61 received a placebo. The amount of caffeine was approximately equal to three cups of coffee.

Motor skills worsened in the caffeine group by an average of 0.16 points on the Movement Disorder Society–sponsored Unified Parkinson’s Disease Rating Scale (MDS-UPDRS) part III during patients’ “on” state and by 0.64 points in the placebo group; the difference was not significant.

The researchers found no differences between groups in the clinical global impression of change based on both patient and examiner assessment. In addition, no differences appeared in depression or anxiety, or in quality of life.

The study findings included long-term follow up data from 88 patients assessed at 12 months and 66 patients assessed at 18 months. The results were similar to the 6-month results, and the study was stopped, although the original design included a 4-year extension.

A total of 29 patients in the caffeine group and 31 patients in the placebo group reported adverse events, and 7 patients in the caffeine group and 5 in the placebo group discontinued the study because of side effects. A serious adverse event was reported by one patient in each group, but neither was deemed related to the interventions.

The findings contrast with the effects of caffeine in the 6-week study in 2012 that showed a significant, 3.2-point improvement in motor skills on the MDS-UPDRS part III with caffeine use, as well as reduced daytime sleepiness, the researchers said (Neurology. 2012;79:651-8). Interpretations of the different findings between the trials may be constrained by factors including differences in the study populations, speed of dose escalation, and trial duration and the possible short-term nature of caffeine’s impact, they noted. “Regardless, our core finding is that caffeine cannot be recommended as symptomatic therapy for parkinsonism.” However, “since caffeine is safe and generally well tolerated, it seems reasonable to empirically try intermittent moderate doses of caffeine for somnolence, and repeat if improvement is seen,” they added.

Dr. Postuma disclosed grant funding from the Canadian Institute of Health Research, the Webster Foundation, and Fonds de Recherche du Québec-Santé for this study.

 

Caffeine consumption has no significant impact on motor skills in patients with Parkinson’s disease, based on data from a double-blind, randomized, placebo-controlled trial of 121 adults. The findings were published online Sept. 27 in Neurology.

Dr. Ronald B. Postuma
In this study, 60 patients received 200 mg of caffeine in capsule form twice daily, in the morning and after lunch for 6 months; 61 received a placebo. The amount of caffeine was approximately equal to three cups of coffee.

Motor skills worsened in the caffeine group by an average of 0.16 points on the Movement Disorder Society–sponsored Unified Parkinson’s Disease Rating Scale (MDS-UPDRS) part III during patients’ “on” state and by 0.64 points in the placebo group; the difference was not significant.

The researchers found no differences between groups in the clinical global impression of change based on both patient and examiner assessment. In addition, no differences appeared in depression or anxiety, or in quality of life.

The study findings included long-term follow up data from 88 patients assessed at 12 months and 66 patients assessed at 18 months. The results were similar to the 6-month results, and the study was stopped, although the original design included a 4-year extension.

A total of 29 patients in the caffeine group and 31 patients in the placebo group reported adverse events, and 7 patients in the caffeine group and 5 in the placebo group discontinued the study because of side effects. A serious adverse event was reported by one patient in each group, but neither was deemed related to the interventions.

The findings contrast with the effects of caffeine in the 6-week study in 2012 that showed a significant, 3.2-point improvement in motor skills on the MDS-UPDRS part III with caffeine use, as well as reduced daytime sleepiness, the researchers said (Neurology. 2012;79:651-8). Interpretations of the different findings between the trials may be constrained by factors including differences in the study populations, speed of dose escalation, and trial duration and the possible short-term nature of caffeine’s impact, they noted. “Regardless, our core finding is that caffeine cannot be recommended as symptomatic therapy for parkinsonism.” However, “since caffeine is safe and generally well tolerated, it seems reasonable to empirically try intermittent moderate doses of caffeine for somnolence, and repeat if improvement is seen,” they added.

Dr. Postuma disclosed grant funding from the Canadian Institute of Health Research, the Webster Foundation, and Fonds de Recherche du Québec-Santé for this study.

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

FROM NEUROLOGY

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

 

Key clinical point: Caffeine consumption showed no benefits in motor symptoms in patients with Parkinson’s disease.

Major finding: Motor skills declined by an average of 0.16 points in the caffeine group and 0.64 points in the placebo group; the difference was not significant.

Data source: A double-blind, multicenter, placebo-controlled trial of 121 adults with Parkinson’s.

Disclosures: Dr. Postuma disclosed grant funding from the Canadian Institute of Health Research, the Webster Foundation, and Fonds de Recherche du Québec-Santé for this study.

Disqus Comments
Default

LUME-Mesa trial: Nintedanib improves PFS in mesothelioma

Article Type
Changed
Fri, 01/04/2019 - 13:41

– Adding the oral kinase inhibitor nintedanib to pemetrexed/cisplatin resulted in substantial improvements in outcomes in patients with unresectable malignant pleural mesothelioma in phase 2 of the randomized, placebo-controlled phase 2/3 LUME-Meso study.

The effects were particularly pronounced among those with epithelioid histology, Jose Barrueco, PhD, of Boehringer Ingelheim Pharmaceuticals, Ridgefield, Conn., reported at the Chicago Multidisciplinary Symposium in Thoracic Oncology.

Progression-free survival – the primary endpoint of the study – was improved in 44 patients randomized to receive up to six cycles of pemetrexed/cisplatin plus nintedanib, compared with 43 patients who received pemetrexed/cisplatin plus placebo (median 9.4 vs. 5.7 months; hazard ratio, 0.54), Dr. Barrueco said.

In the 89% of patients with epithelioid malignant pleural mesothelioma, progression-free survival was a median of 9.7 vs. 5.7 months with nintedanib vs. placebo (HR, 0.49).

There was a trend toward improved overall survival in the nintedanib group vs. the placebo group, (median 18.3 vs. 14.2 months; HR, 0.77; P = .319), and overall survival was slightly better in those with epithelioid histology (median 20.6 vs. 15.2 months ; HR, 0.70; P = .197).

Consistent with these results, the adjusted mean change in forced vital capacity at cycle eight also favored nintedanib over placebo (+10.0 vs. +2.8 for a mean treatment difference of 7.2 overall, and +14.1 vs. +4.2 for a mean treatment difference of 9.9 in those with epithelioid histology).

“Overall frequency of adverse events was consistent with the known safety profile of nintedanib,” Dr. Barrueco said, noting that most adverse events were reversible with dose reduction.

Study participants were chemotherapy-naive patients with a mean age of 67 years and Eastern Cooperative Oncology Group performance status of 0-1. They received pemetrexed at a dose of 500 mg/m2 and cisplatin at a dose of 75 mg/m2 on day 1 plus either nintedanib at a dose of 200 mg twice daily on days 2-21 or placebo, followed by monotherapy with nintedanib or placebo until progression or unacceptable toxicity.

“In conclusion, nintedanib plus pemetrexed/cisplatin demonstrated a signal for clinical benefit in the first-time treatment of patients with malignant pleural mesothelioma. This was evident in all endpoints of the trial, and consistently showed benefit for the nintedanib group,” Mr. Barrueco said, noting that phase 3 of the LUME-Meso study is now recruiting.

Meeting/Event
Publications
Topics
Sections
Meeting/Event
Meeting/Event
Related Articles

– Adding the oral kinase inhibitor nintedanib to pemetrexed/cisplatin resulted in substantial improvements in outcomes in patients with unresectable malignant pleural mesothelioma in phase 2 of the randomized, placebo-controlled phase 2/3 LUME-Meso study.

The effects were particularly pronounced among those with epithelioid histology, Jose Barrueco, PhD, of Boehringer Ingelheim Pharmaceuticals, Ridgefield, Conn., reported at the Chicago Multidisciplinary Symposium in Thoracic Oncology.

Progression-free survival – the primary endpoint of the study – was improved in 44 patients randomized to receive up to six cycles of pemetrexed/cisplatin plus nintedanib, compared with 43 patients who received pemetrexed/cisplatin plus placebo (median 9.4 vs. 5.7 months; hazard ratio, 0.54), Dr. Barrueco said.

In the 89% of patients with epithelioid malignant pleural mesothelioma, progression-free survival was a median of 9.7 vs. 5.7 months with nintedanib vs. placebo (HR, 0.49).

There was a trend toward improved overall survival in the nintedanib group vs. the placebo group, (median 18.3 vs. 14.2 months; HR, 0.77; P = .319), and overall survival was slightly better in those with epithelioid histology (median 20.6 vs. 15.2 months ; HR, 0.70; P = .197).

Consistent with these results, the adjusted mean change in forced vital capacity at cycle eight also favored nintedanib over placebo (+10.0 vs. +2.8 for a mean treatment difference of 7.2 overall, and +14.1 vs. +4.2 for a mean treatment difference of 9.9 in those with epithelioid histology).

“Overall frequency of adverse events was consistent with the known safety profile of nintedanib,” Dr. Barrueco said, noting that most adverse events were reversible with dose reduction.

Study participants were chemotherapy-naive patients with a mean age of 67 years and Eastern Cooperative Oncology Group performance status of 0-1. They received pemetrexed at a dose of 500 mg/m2 and cisplatin at a dose of 75 mg/m2 on day 1 plus either nintedanib at a dose of 200 mg twice daily on days 2-21 or placebo, followed by monotherapy with nintedanib or placebo until progression or unacceptable toxicity.

“In conclusion, nintedanib plus pemetrexed/cisplatin demonstrated a signal for clinical benefit in the first-time treatment of patients with malignant pleural mesothelioma. This was evident in all endpoints of the trial, and consistently showed benefit for the nintedanib group,” Mr. Barrueco said, noting that phase 3 of the LUME-Meso study is now recruiting.

– Adding the oral kinase inhibitor nintedanib to pemetrexed/cisplatin resulted in substantial improvements in outcomes in patients with unresectable malignant pleural mesothelioma in phase 2 of the randomized, placebo-controlled phase 2/3 LUME-Meso study.

The effects were particularly pronounced among those with epithelioid histology, Jose Barrueco, PhD, of Boehringer Ingelheim Pharmaceuticals, Ridgefield, Conn., reported at the Chicago Multidisciplinary Symposium in Thoracic Oncology.

Progression-free survival – the primary endpoint of the study – was improved in 44 patients randomized to receive up to six cycles of pemetrexed/cisplatin plus nintedanib, compared with 43 patients who received pemetrexed/cisplatin plus placebo (median 9.4 vs. 5.7 months; hazard ratio, 0.54), Dr. Barrueco said.

In the 89% of patients with epithelioid malignant pleural mesothelioma, progression-free survival was a median of 9.7 vs. 5.7 months with nintedanib vs. placebo (HR, 0.49).

There was a trend toward improved overall survival in the nintedanib group vs. the placebo group, (median 18.3 vs. 14.2 months; HR, 0.77; P = .319), and overall survival was slightly better in those with epithelioid histology (median 20.6 vs. 15.2 months ; HR, 0.70; P = .197).

Consistent with these results, the adjusted mean change in forced vital capacity at cycle eight also favored nintedanib over placebo (+10.0 vs. +2.8 for a mean treatment difference of 7.2 overall, and +14.1 vs. +4.2 for a mean treatment difference of 9.9 in those with epithelioid histology).

“Overall frequency of adverse events was consistent with the known safety profile of nintedanib,” Dr. Barrueco said, noting that most adverse events were reversible with dose reduction.

Study participants were chemotherapy-naive patients with a mean age of 67 years and Eastern Cooperative Oncology Group performance status of 0-1. They received pemetrexed at a dose of 500 mg/m2 and cisplatin at a dose of 75 mg/m2 on day 1 plus either nintedanib at a dose of 200 mg twice daily on days 2-21 or placebo, followed by monotherapy with nintedanib or placebo until progression or unacceptable toxicity.

“In conclusion, nintedanib plus pemetrexed/cisplatin demonstrated a signal for clinical benefit in the first-time treatment of patients with malignant pleural mesothelioma. This was evident in all endpoints of the trial, and consistently showed benefit for the nintedanib group,” Mr. Barrueco said, noting that phase 3 of the LUME-Meso study is now recruiting.

Publications
Publications
Topics
Article Type
Sections
Article Source

AT A SYMPOSIUM IN THORACIC ONCOLOGY

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

 

Key clinical point: Adding nintedanib to pemetrexed/cisplatin resulted in improved PFS in patients with mesothelioma in phase 2 of the LUME-Meso study.

Major finding: Overall median PFS with nintedanib vs. placebo was 9.4 vs. 5.7 months (HR, 0.54).

Data source: Phase 2 of the LUME-Meso trial with 87 patients.

Disclosures: Dr. Barrueco is an employee of Boehringer Ingelheim.

Disqus Comments
Default

Risk of Osteoporotic Fracture After Steroid Injections in Patients With Medicare

Article Type
Changed
Thu, 09/19/2019 - 13:20

Take-Home Points

  • Analysis of patients in the Medicare database showed that each successive ESI decreased the risk of an osteoporotic spine fracture by 2%, and that each successive LJSI decreases it by 4%.

  • Although statistically significant, this may not be clinically relevant.

  • Successive ESI did not influence the risk of developing an osteoporotic hip or wrist fracture, but that each additional LJSI reduced the risk.

  • Prolonged steroid exposure was found to increase the risk of spine fracture for ESI and LJSI patients.

  • Acute exposure to exogenous steroids via the epidural space, transforaminal space, or large joints does not seem to increase the risk of an osteoporotic fracture of the spine, hip, or wrist.

Epidural steroid injections (ESIs) are widely used in the nonoperative treatment of low back pain, radicular leg pain, and spinal stenosis. The treatment rationale is that locally injected anti-inflammatory drugs, such as steroids, reduce inflammation by inhibiting formation and release of inflammatory cytokines, leading to pain reduction.1,2 According to 4 systematic reviews, the best available evidence of the efficacy of ESIs is less than robust.3-6 These reviews were limited by the heterogeneity of patient selection, delivery mode, type and dose of steroid used, number and frequency of ESIs, and outcome measures.

The association of chronic oral steroid use and the development of osteoporosis was previously established.7,8 One concern is that acute exposure to steroids in the form of lumbar ESIs may also lead to osteoporosis and then a pathologic fracture of the vertebra. Several studies have found no association between bone mineral density and cumulative steroid dose,9,10 mean number of ESIs, or duration of ESIs,10 though other studies have found lower bone mineral density in postmenopausal women treated with ESIs.11-13

In a study of 3000 ESI patients propensity-matched to a non-ESI cohort, Mandel and colleagues14 found that each successive ESI increased the risk of osteoporotic spine fracture by 21%. This clinically relevant 21% increased risk might lead physicians to stop prescribing or using this intervention. However, the association between osteoporotic fractures and other types of steroid injections remains poorly understood and underinvestigated. 

To further evaluate the relationship between steroid injections and osteoporotic fracture risk, we analyzed Medicare administrative claims data on both large-joint steroid injections (LJSIs) into knee and hip and transforaminal steroid injections (TSIs), as well as osteoporotic hip and wrist fractures. Our hypothesis was that a systemic effect of steroid injections would increase fracture risk in all skeletal locations regardless of injection site, whereas a local effect would produce a disproportionate increased risk of spine fracture with spine injection.

Materials and Methods

Medicare is a publicly funded US health insurance program for people 65 years old or older, people under age 65 years with certain disabilities, and people (any age) with end-stage renal disease or amyotrophic lateral sclerosis. The 5% Medicare Part B (physician, carrier) dataset contains individual claims records for a random sample of Medicare beneficiaries (~2.4 million enrollees). Patients who received steroid injections were identified from 5% Medicare claims made between January 1, 2004 and December 31, 2011. LJSIs were identified by Current Procedural Terminology (CPT) code 20610 and any of 16 other CPT codes: J0702, J1020, J1030, J1040, J1094, J1100, J1700, J1710, J1720, J2650, J2920, J2930, J3300, J3301, J3302, and J3303. ESIs were identified by CPT code 62310, 62311, 62318, or 62319, and TSIs by CPT code 64479, 64480, 64483, or 64484. Patients were followed in their initial injection cohort. For example, a patient who received an ESI initially and later received an LJSI remained in the ESI cohort.

Several groups of patients were excluded from the study: those who received Medicare coverage because of their age (under 65 years) and disabilities; those who received Medicare health benefits through health maintenance organizations (healthcare expenses were not submitted to the Centers for Medicare & Medicaid Services for payment, and therefore claims were not in the database or were incomplete); those with a prior claim history of <12 months (incomplete comorbidity history); and those who received a diagnosis of osteoporotic fracture (International Classification of Diseases, Ninth Revision, Clinical Modification [ICD-9-CM] code 733.1x) before the initial steroid injection.

We determined the incidence of osteoporotic wrist, hip, and spine fractures within 1, 2, and 8 years after LJSI, ESI, and TSI. Wrist, hip, and spine fractures were identified by ICD-9-CM diagnosis codes 733.12, 733.13, and 733.14, respectively. We also determined the number of steroid injections given before wrist, hip, or spine fracture or, if no fracture occurred, before death or the end of the data period.

Statistical Analysis

Multivariate Cox regression analysis was performed to evaluate the risk factors for wrist, spine, and hip fractures. The covariates in this model included age, sex, race, census region, Medicare buy-in status, Charlson Comorbidity Index (CCI),15 year, and number of steroid injections before fracture, death, or end of data period. Medicare buy-in status, which indicates whether the beneficiary received financial assistance in paying insurance premiums, was used as a proxy for socioeconomic status. CCI is used as a composite score of a patient’s general health status in terms of comorbidities.15,16 Four previously established categories17 were used to group CCIs in this study: 0 (none), 1 to 2 (low), 3 to 4 (moderate), and 5 or more (high). In addition, several diagnoses made within the 12 months before initial steroid injection were considered: osteoporosis (ICD-9-CM codes 733.0x, V82.81), Cushing syndrome (ICD-9-CM code 255.0), long-term (current) use of bisphosphonates (ICD-9-CM code V58.68), asymptomatic postmenopausal status (ICD-9-CM code V49.81), postmenopausal hormone replacement therapy (ICD-9-CM code V07.4), and long-term (current) use of steroids (ICD-9-CM code V58.65). The comparison of relative risk between any groups was reported as the adjusted hazard ratio (AHR), which is the ratio of the hazard rates of that particular outcome, taking into account inherent patient characteristics such as age, sex, and race as covariates. AHR of 1 corresponds to equivalent risk, AHR of >1 to elevated risk, and AHR of <1 to reduced risk.

Results

Using the 5% Medicare data for 2004 to 2011, we identified 275,999 Medicare beneficiaries who underwent LJSI, 93,943 who underwent ESI, and 32,311 who underwent TSI. During this period, TSI use increased, ESI use decreased, and LJSI use was relatively stable (Figure).

Figure.
LJSI patients underwent a mean (SD) of 3.0 (4.1) LJSIs; ESI patients, 3.2 (3.9) ESIs; and TSI patients, 7.5 (12.1) TSIs. Very few patients had more than 1 steroid injection site (Table 1).
Table 1.

The risk for osteoporotic spine fracture 1, 2, and 8 years after ESI, TSI, or LJSI was affected by age, race, sex, and CCI (P < .001 for all; Tables 2-4).

Table 2.
Patients with higher CCIs, females, older patients, and white patients had higher spine fracture risks. For ESI and LJSI patients, long-term steroid use was also associated with higher spine fracture risk at all examined time points.
Table 3.
For example, 8 years after ESI and LJSI, patients with long-term steroid use had 88% (P = .002) and 47% (P = .008) elevated risk of spine fracture, respectively. For TSI patients, spine fracture risk tended to be higher in patients with long-term steroid use 1 and 2 years after injection; this association was significant 8 years after injection (P = .046). ESI patients of lower socioeconomic status (Medicare buy-in) also had higher spine fracture risk at all examined time points, whereas LJSI and TSI patients did not. Two years after injection, each additional ESI decreased the relative risk of spine fracture by 2% (P < .001), each additional TSI decreased it by 1% (P = .009), and each additional LJSI decreased it by 4% (P < .001). 
Table 4.

The risk for osteoporotic hip fracture after 1 and 2 years was affected by age and number of LJSIs and TSIs but not by number of ESIs. Sex and CCI were also risk factors for hip fracture at 1 and 2 years for ESI and LJSI patients, as was race for LJSI patients. Risk for osteoporotic wrist fracture at 1 and 2 years was affected by sex and race for ESI and LJSI patients; age, race, CCI, and long-term steroid use were risk factors for TSI patients at all time points. Higher number of LJSIs, but not ESIs or TSIs, was associated with lower wrist fracture risk.

Discussion

ESIs continue to be used in the nonoperative treatment of low back pain, radicular leg pain, and spinal stenosis. Although the present study found ESI use increased in the Medicare population between 1994 and 2001,18 the trend is reversing, decreasing by 25%, with rates of 264 per 10,000 Medicare enrollees in 2004 and 194 per 10,000 enrollees in 2011. ESI use may have changed after systematic reviews revealed there was no clear evidence of the efficacy of ESIs in managing low back pain and radicular leg pain3,5,6 or spinal stenosis.4

Nevertheless, ESIs are widely used because of the perceived benefit balanced against the perceived rarity of adverse events.6 Even if patients recognize a low likelihood of significant benefit, they may accept ESI as preferable to surgery. In addition, most private payers require extensive nonoperative treatment before they will approve surgery as a treatment option.

In a study by Mandel and colleagues,14 ESI increased the risk of vertebral compression fractures by 21%, which in turn increased the risk of death.19 If accurate, these findings obviously would challenge the perception that ESI is a low-risk intervention. In contrast to the Mandel study,14 the present analysis of the Medicare population revealed no clinically relevant change in risk of osteoporotic spine fracture with each successive ESI after the initial injection. After the initial injection, each successive ESI decreased the relative risk of osteoporotic spine fracture by 2%, and each successive LJSI decreased it by 4%. Although statistically significant, the small change in relative risk may not be clinically relevant. However, taken cumulatively over a number of successive injections, these effects may be clinically relevant.

The data also showed that, after the initial injection, each successive ESI had no effect on risk of osteoporotic hip or wrist fracture, and each successive LJSI reduced the risk. Similar to earlier findings,20,21 long-term steroid use increased the risk of spine fracture in ESI and LJSI patients. Prolonged exposure to steroids may be necessary to reduce bone formation and increase bone breakdown.12

Although the study by Mandel and colleagues14 and our study both used administrative databases and survival analysis methods, conclusions differed. First, Mandel and colleagues14 used a study inclusion criterion of spine-related steroid injections, whereas we used a criterion of any steroid injection. Second, they used 50 years as the lower age for study inclusion, and we used 65 years. Third, to control for patients who had osteoporosis before study entry, they excluded those who had a fracture in an adjacent vertebra after kyphoplasty and vertebroplasty. It is unclear if patients who had osteoporotic fractures at other sites were excluded as well. Thus, the 2 cohorts may not be directly comparable.

Whereas Mandel and colleagues14 based their definition of osteoporotic spine fracture on a keyword search of a radiology database, we used a specific reportable ICD-9-CM diagnosis code. As a result, they may have overreported osteoporotic spine fractures, and we may have underreported. Finally, our sample was much larger than theirs. Given the relative rarity of osteoporotic fractures, a study with a larger sample may have more power to detect differences. In addition, unlike Mandel and colleagues,14 we focused on an injection cohort. We did not include or make comparisons with a no-injection cohort because our study hypothesis involved the potential systemic effects of steroid injections based on injection site. Although chronic steroid use was found to have a significant effect in our study, it is unclear to what extent the diagnosis code was used, during the comorbidity assessment or only in the event of steroid-related complications.

Our study also found that, after the initial injection, each successive LJSI decreased the risk of osteoporotic wrist fracture by 10%, and each successive TSI decreased the risk of osteoporotic hip fracture by 5%. It is plausible these injections allowed improved mobility, mitigating the effects of osteoporosis induced by inactivity and lack of resistance training. It is also possible that improved mobility limited falls.

In summary, this analysis of the Medicare claims database revealed that ESI, TSI, and LJSI decreased osteoporotic spine fracture risk. However, the effect was small and may not be clinically meaningful. After the initial injection, successive ESIs had no effect on the risk of osteoporotic hip or wrist fracture, and successive LJSIs reduced the risk of osteoporotic wrist fracture, perhaps because of improved mobility. Prolonged oral steroid use increased spine fracture risk in ESI and LJSI patients. More studies are needed to evaluate the risk-benefit profile of steroid injections.

References

1. Pethö G, Reeh PW. Sensory and signaling mechanisms of bradykinin, eicosanoids, platelet-activating factor, and nitric oxide in peripheral nociceptors. Physiol Rev. 2012;92(4):1699-1775.

2. Saal J. The role of inflammation in lumbar pain. Spine. 1995;20(16):1821-1827.

3. Choi HJ, Hahn S, Kim CH, et al. Epidural steroid injection therapy for low back pain: a meta-analysis. Int J Technol Assess Health Care. 2013;29(3):244-253.

4. Chou R, Loeser JD, Owens DK, et al; American Pain Society Low Back Pain Guideline Panel. Interventional therapies, surgery, and interdisciplinary rehabilitation for low back pain: an evidence-based clinical practice guideline from the American Pain Society. Spine. 2009;34(10):1066-1077. 

5. Savigny P, Watson P, Underwood M; Guideline Development Group. Early management of persistent non-specific low back pain: summary of NICE guidance. BMJ. 2009;338:b1805.

6. Staal JB, de Bie RA, de Vet HC, Hildebrandt J, Nelemans P. Injection therapy for subacute and chronic low back pain: an updated Cochrane review. Spine. 2009;34(1):49-59.

7. Angeli A, Guglielmi G, Dovio A, et al. High prevalence of asymptomatic vertebral fractures in post-menopausal women receiving chronic glucocorticoid therapy: a cross-sectional outpatient study. Bone. 2006;39(2):253-259.

8. Donnan PT, Libby G, Boyter AC, Thompson P. The population risk of fractures attributable to oral corticosteroids. Pharmacoepidemiol Drug Saf. 2005;14(3):177-186.

9. Dubois EF, Wagemans MF, Verdouw BC, et al. Lack of relationships between cumulative methylprednisolone dose and bone mineral density in healthy men and postmenopausal women with chronic low back pain. Clin Rheumatol. 2003;22(1):12-17.

10. Yi Y, Hwang B, Son H, Cheong I. Low bone mineral density, but not epidural steroid injection, is associated with fracture in postmenopausal women with low back pain. Pain Physician. 2012;15(6):441-449. 

11. Al-Shoha A, Rao DS, Schilling J, Peterson E, Mandel S. Effect of epidural steroid injection on bone mineral density and markers of bone turnover in postmenopausal women. Spine. 2012;37(25):E1567-E1571. 

12. Kang SS, Hwang BM, Son H, Cheong IY, Lee SJ, Chung TY. Changes in bone mineral density in postmenopausal women treated with epidural steroid injections for lower back pain. Pain Physician. 2012;15(3):229-236.

13. Kim S, Hwang B. Relationship between bone mineral density and the frequent administration of epidural steroid injections in postmenopausal women with low back pain. Pain Res Manag. 2014;19(1):30-34. 

14. Mandel S, Schilling J, Peterson E, Rao DS, Sanders W. A retrospective analysis of vertebral body fractures following epidural steroid injections. J Bone Joint Surg Am. 2013;95(11):961-964. 

15. Charlson ME, Pompei P, Ales KL, MacKenzie CR. A new method of classifying prognostic comorbidity in longitudinal studies: development and validation. J Chronic Dis. 1987;40(5):373-383.

16. Deyo RA, Cherkin DC, Ciol MA. Adapting a clinical comorbidity index for use with ICD-9-CM administrative databases. J Clin Epidemiol. 1992;45(6):613-619.

17. Murray SB, Bates DW, Ngo L, Ufberg JW, Shapiro NI. Charlson index is associated with one-year mortality in emergency department patients with suspected infection. Acad Emerg Med. 2006;13(5):530-536.

18. Friedly J, Chan L, Deyo R. Increases in lumbosacral injections in the Medicare population: 1994 to 2001. Spine. 2007;32(16):1754-1760.

19. Puisto V, Rissanen H, Heliövaara M, et al. Vertebral fracture and cause-specific mortality: a prospective population study of 3,210 men and 3,730 women with 30 years of follow-up. Eur Spine J. 2011;20(12):2181-2186. 

20. Lee YH, Woo JH, Choi SJ, Ji JD, Song GG. Effects of low-dose corticosteroids on the bone mineral density of patients with rheumatoid arthritis: a meta-analysis. J Investig Med. 2008;56(8):1011-1018.

21. Lukert BP, Raisz LG. Glucocorticoid-induced osteoporosis. Rheum Dis Clin North Am. 1994;20(3):629-650.

Article PDF
Author and Disclosure Information

Authors' Disclosure Statement: The authors report no actual or potential conflict of interest in relation to this article. 

Issue
The American Journal of Orthopedics - 46(5)
Publications
Topics
Page Number
E293-E300
Sections
Author and Disclosure Information

Authors' Disclosure Statement: The authors report no actual or potential conflict of interest in relation to this article. 

Author and Disclosure Information

Authors' Disclosure Statement: The authors report no actual or potential conflict of interest in relation to this article. 

Article PDF
Article PDF

Take-Home Points

  • Analysis of patients in the Medicare database showed that each successive ESI decreased the risk of an osteoporotic spine fracture by 2%, and that each successive LJSI decreases it by 4%.

  • Although statistically significant, this may not be clinically relevant.

  • Successive ESI did not influence the risk of developing an osteoporotic hip or wrist fracture, but that each additional LJSI reduced the risk.

  • Prolonged steroid exposure was found to increase the risk of spine fracture for ESI and LJSI patients.

  • Acute exposure to exogenous steroids via the epidural space, transforaminal space, or large joints does not seem to increase the risk of an osteoporotic fracture of the spine, hip, or wrist.

Epidural steroid injections (ESIs) are widely used in the nonoperative treatment of low back pain, radicular leg pain, and spinal stenosis. The treatment rationale is that locally injected anti-inflammatory drugs, such as steroids, reduce inflammation by inhibiting formation and release of inflammatory cytokines, leading to pain reduction.1,2 According to 4 systematic reviews, the best available evidence of the efficacy of ESIs is less than robust.3-6 These reviews were limited by the heterogeneity of patient selection, delivery mode, type and dose of steroid used, number and frequency of ESIs, and outcome measures.

The association of chronic oral steroid use and the development of osteoporosis was previously established.7,8 One concern is that acute exposure to steroids in the form of lumbar ESIs may also lead to osteoporosis and then a pathologic fracture of the vertebra. Several studies have found no association between bone mineral density and cumulative steroid dose,9,10 mean number of ESIs, or duration of ESIs,10 though other studies have found lower bone mineral density in postmenopausal women treated with ESIs.11-13

In a study of 3000 ESI patients propensity-matched to a non-ESI cohort, Mandel and colleagues14 found that each successive ESI increased the risk of osteoporotic spine fracture by 21%. This clinically relevant 21% increased risk might lead physicians to stop prescribing or using this intervention. However, the association between osteoporotic fractures and other types of steroid injections remains poorly understood and underinvestigated. 

To further evaluate the relationship between steroid injections and osteoporotic fracture risk, we analyzed Medicare administrative claims data on both large-joint steroid injections (LJSIs) into knee and hip and transforaminal steroid injections (TSIs), as well as osteoporotic hip and wrist fractures. Our hypothesis was that a systemic effect of steroid injections would increase fracture risk in all skeletal locations regardless of injection site, whereas a local effect would produce a disproportionate increased risk of spine fracture with spine injection.

Materials and Methods

Medicare is a publicly funded US health insurance program for people 65 years old or older, people under age 65 years with certain disabilities, and people (any age) with end-stage renal disease or amyotrophic lateral sclerosis. The 5% Medicare Part B (physician, carrier) dataset contains individual claims records for a random sample of Medicare beneficiaries (~2.4 million enrollees). Patients who received steroid injections were identified from 5% Medicare claims made between January 1, 2004 and December 31, 2011. LJSIs were identified by Current Procedural Terminology (CPT) code 20610 and any of 16 other CPT codes: J0702, J1020, J1030, J1040, J1094, J1100, J1700, J1710, J1720, J2650, J2920, J2930, J3300, J3301, J3302, and J3303. ESIs were identified by CPT code 62310, 62311, 62318, or 62319, and TSIs by CPT code 64479, 64480, 64483, or 64484. Patients were followed in their initial injection cohort. For example, a patient who received an ESI initially and later received an LJSI remained in the ESI cohort.

Several groups of patients were excluded from the study: those who received Medicare coverage because of their age (under 65 years) and disabilities; those who received Medicare health benefits through health maintenance organizations (healthcare expenses were not submitted to the Centers for Medicare & Medicaid Services for payment, and therefore claims were not in the database or were incomplete); those with a prior claim history of <12 months (incomplete comorbidity history); and those who received a diagnosis of osteoporotic fracture (International Classification of Diseases, Ninth Revision, Clinical Modification [ICD-9-CM] code 733.1x) before the initial steroid injection.

We determined the incidence of osteoporotic wrist, hip, and spine fractures within 1, 2, and 8 years after LJSI, ESI, and TSI. Wrist, hip, and spine fractures were identified by ICD-9-CM diagnosis codes 733.12, 733.13, and 733.14, respectively. We also determined the number of steroid injections given before wrist, hip, or spine fracture or, if no fracture occurred, before death or the end of the data period.

Statistical Analysis

Multivariate Cox regression analysis was performed to evaluate the risk factors for wrist, spine, and hip fractures. The covariates in this model included age, sex, race, census region, Medicare buy-in status, Charlson Comorbidity Index (CCI),15 year, and number of steroid injections before fracture, death, or end of data period. Medicare buy-in status, which indicates whether the beneficiary received financial assistance in paying insurance premiums, was used as a proxy for socioeconomic status. CCI is used as a composite score of a patient’s general health status in terms of comorbidities.15,16 Four previously established categories17 were used to group CCIs in this study: 0 (none), 1 to 2 (low), 3 to 4 (moderate), and 5 or more (high). In addition, several diagnoses made within the 12 months before initial steroid injection were considered: osteoporosis (ICD-9-CM codes 733.0x, V82.81), Cushing syndrome (ICD-9-CM code 255.0), long-term (current) use of bisphosphonates (ICD-9-CM code V58.68), asymptomatic postmenopausal status (ICD-9-CM code V49.81), postmenopausal hormone replacement therapy (ICD-9-CM code V07.4), and long-term (current) use of steroids (ICD-9-CM code V58.65). The comparison of relative risk between any groups was reported as the adjusted hazard ratio (AHR), which is the ratio of the hazard rates of that particular outcome, taking into account inherent patient characteristics such as age, sex, and race as covariates. AHR of 1 corresponds to equivalent risk, AHR of >1 to elevated risk, and AHR of <1 to reduced risk.

Results

Using the 5% Medicare data for 2004 to 2011, we identified 275,999 Medicare beneficiaries who underwent LJSI, 93,943 who underwent ESI, and 32,311 who underwent TSI. During this period, TSI use increased, ESI use decreased, and LJSI use was relatively stable (Figure).

Figure.
LJSI patients underwent a mean (SD) of 3.0 (4.1) LJSIs; ESI patients, 3.2 (3.9) ESIs; and TSI patients, 7.5 (12.1) TSIs. Very few patients had more than 1 steroid injection site (Table 1).
Table 1.

The risk for osteoporotic spine fracture 1, 2, and 8 years after ESI, TSI, or LJSI was affected by age, race, sex, and CCI (P < .001 for all; Tables 2-4).

Table 2.
Patients with higher CCIs, females, older patients, and white patients had higher spine fracture risks. For ESI and LJSI patients, long-term steroid use was also associated with higher spine fracture risk at all examined time points.
Table 3.
For example, 8 years after ESI and LJSI, patients with long-term steroid use had 88% (P = .002) and 47% (P = .008) elevated risk of spine fracture, respectively. For TSI patients, spine fracture risk tended to be higher in patients with long-term steroid use 1 and 2 years after injection; this association was significant 8 years after injection (P = .046). ESI patients of lower socioeconomic status (Medicare buy-in) also had higher spine fracture risk at all examined time points, whereas LJSI and TSI patients did not. Two years after injection, each additional ESI decreased the relative risk of spine fracture by 2% (P < .001), each additional TSI decreased it by 1% (P = .009), and each additional LJSI decreased it by 4% (P < .001). 
Table 4.

The risk for osteoporotic hip fracture after 1 and 2 years was affected by age and number of LJSIs and TSIs but not by number of ESIs. Sex and CCI were also risk factors for hip fracture at 1 and 2 years for ESI and LJSI patients, as was race for LJSI patients. Risk for osteoporotic wrist fracture at 1 and 2 years was affected by sex and race for ESI and LJSI patients; age, race, CCI, and long-term steroid use were risk factors for TSI patients at all time points. Higher number of LJSIs, but not ESIs or TSIs, was associated with lower wrist fracture risk.

Discussion

ESIs continue to be used in the nonoperative treatment of low back pain, radicular leg pain, and spinal stenosis. Although the present study found ESI use increased in the Medicare population between 1994 and 2001,18 the trend is reversing, decreasing by 25%, with rates of 264 per 10,000 Medicare enrollees in 2004 and 194 per 10,000 enrollees in 2011. ESI use may have changed after systematic reviews revealed there was no clear evidence of the efficacy of ESIs in managing low back pain and radicular leg pain3,5,6 or spinal stenosis.4

Nevertheless, ESIs are widely used because of the perceived benefit balanced against the perceived rarity of adverse events.6 Even if patients recognize a low likelihood of significant benefit, they may accept ESI as preferable to surgery. In addition, most private payers require extensive nonoperative treatment before they will approve surgery as a treatment option.

In a study by Mandel and colleagues,14 ESI increased the risk of vertebral compression fractures by 21%, which in turn increased the risk of death.19 If accurate, these findings obviously would challenge the perception that ESI is a low-risk intervention. In contrast to the Mandel study,14 the present analysis of the Medicare population revealed no clinically relevant change in risk of osteoporotic spine fracture with each successive ESI after the initial injection. After the initial injection, each successive ESI decreased the relative risk of osteoporotic spine fracture by 2%, and each successive LJSI decreased it by 4%. Although statistically significant, the small change in relative risk may not be clinically relevant. However, taken cumulatively over a number of successive injections, these effects may be clinically relevant.

The data also showed that, after the initial injection, each successive ESI had no effect on risk of osteoporotic hip or wrist fracture, and each successive LJSI reduced the risk. Similar to earlier findings,20,21 long-term steroid use increased the risk of spine fracture in ESI and LJSI patients. Prolonged exposure to steroids may be necessary to reduce bone formation and increase bone breakdown.12

Although the study by Mandel and colleagues14 and our study both used administrative databases and survival analysis methods, conclusions differed. First, Mandel and colleagues14 used a study inclusion criterion of spine-related steroid injections, whereas we used a criterion of any steroid injection. Second, they used 50 years as the lower age for study inclusion, and we used 65 years. Third, to control for patients who had osteoporosis before study entry, they excluded those who had a fracture in an adjacent vertebra after kyphoplasty and vertebroplasty. It is unclear if patients who had osteoporotic fractures at other sites were excluded as well. Thus, the 2 cohorts may not be directly comparable.

Whereas Mandel and colleagues14 based their definition of osteoporotic spine fracture on a keyword search of a radiology database, we used a specific reportable ICD-9-CM diagnosis code. As a result, they may have overreported osteoporotic spine fractures, and we may have underreported. Finally, our sample was much larger than theirs. Given the relative rarity of osteoporotic fractures, a study with a larger sample may have more power to detect differences. In addition, unlike Mandel and colleagues,14 we focused on an injection cohort. We did not include or make comparisons with a no-injection cohort because our study hypothesis involved the potential systemic effects of steroid injections based on injection site. Although chronic steroid use was found to have a significant effect in our study, it is unclear to what extent the diagnosis code was used, during the comorbidity assessment or only in the event of steroid-related complications.

Our study also found that, after the initial injection, each successive LJSI decreased the risk of osteoporotic wrist fracture by 10%, and each successive TSI decreased the risk of osteoporotic hip fracture by 5%. It is plausible these injections allowed improved mobility, mitigating the effects of osteoporosis induced by inactivity and lack of resistance training. It is also possible that improved mobility limited falls.

In summary, this analysis of the Medicare claims database revealed that ESI, TSI, and LJSI decreased osteoporotic spine fracture risk. However, the effect was small and may not be clinically meaningful. After the initial injection, successive ESIs had no effect on the risk of osteoporotic hip or wrist fracture, and successive LJSIs reduced the risk of osteoporotic wrist fracture, perhaps because of improved mobility. Prolonged oral steroid use increased spine fracture risk in ESI and LJSI patients. More studies are needed to evaluate the risk-benefit profile of steroid injections.

Take-Home Points

  • Analysis of patients in the Medicare database showed that each successive ESI decreased the risk of an osteoporotic spine fracture by 2%, and that each successive LJSI decreases it by 4%.

  • Although statistically significant, this may not be clinically relevant.

  • Successive ESI did not influence the risk of developing an osteoporotic hip or wrist fracture, but that each additional LJSI reduced the risk.

  • Prolonged steroid exposure was found to increase the risk of spine fracture for ESI and LJSI patients.

  • Acute exposure to exogenous steroids via the epidural space, transforaminal space, or large joints does not seem to increase the risk of an osteoporotic fracture of the spine, hip, or wrist.

Epidural steroid injections (ESIs) are widely used in the nonoperative treatment of low back pain, radicular leg pain, and spinal stenosis. The treatment rationale is that locally injected anti-inflammatory drugs, such as steroids, reduce inflammation by inhibiting formation and release of inflammatory cytokines, leading to pain reduction.1,2 According to 4 systematic reviews, the best available evidence of the efficacy of ESIs is less than robust.3-6 These reviews were limited by the heterogeneity of patient selection, delivery mode, type and dose of steroid used, number and frequency of ESIs, and outcome measures.

The association of chronic oral steroid use and the development of osteoporosis was previously established.7,8 One concern is that acute exposure to steroids in the form of lumbar ESIs may also lead to osteoporosis and then a pathologic fracture of the vertebra. Several studies have found no association between bone mineral density and cumulative steroid dose,9,10 mean number of ESIs, or duration of ESIs,10 though other studies have found lower bone mineral density in postmenopausal women treated with ESIs.11-13

In a study of 3000 ESI patients propensity-matched to a non-ESI cohort, Mandel and colleagues14 found that each successive ESI increased the risk of osteoporotic spine fracture by 21%. This clinically relevant 21% increased risk might lead physicians to stop prescribing or using this intervention. However, the association between osteoporotic fractures and other types of steroid injections remains poorly understood and underinvestigated. 

To further evaluate the relationship between steroid injections and osteoporotic fracture risk, we analyzed Medicare administrative claims data on both large-joint steroid injections (LJSIs) into knee and hip and transforaminal steroid injections (TSIs), as well as osteoporotic hip and wrist fractures. Our hypothesis was that a systemic effect of steroid injections would increase fracture risk in all skeletal locations regardless of injection site, whereas a local effect would produce a disproportionate increased risk of spine fracture with spine injection.

Materials and Methods

Medicare is a publicly funded US health insurance program for people 65 years old or older, people under age 65 years with certain disabilities, and people (any age) with end-stage renal disease or amyotrophic lateral sclerosis. The 5% Medicare Part B (physician, carrier) dataset contains individual claims records for a random sample of Medicare beneficiaries (~2.4 million enrollees). Patients who received steroid injections were identified from 5% Medicare claims made between January 1, 2004 and December 31, 2011. LJSIs were identified by Current Procedural Terminology (CPT) code 20610 and any of 16 other CPT codes: J0702, J1020, J1030, J1040, J1094, J1100, J1700, J1710, J1720, J2650, J2920, J2930, J3300, J3301, J3302, and J3303. ESIs were identified by CPT code 62310, 62311, 62318, or 62319, and TSIs by CPT code 64479, 64480, 64483, or 64484. Patients were followed in their initial injection cohort. For example, a patient who received an ESI initially and later received an LJSI remained in the ESI cohort.

Several groups of patients were excluded from the study: those who received Medicare coverage because of their age (under 65 years) and disabilities; those who received Medicare health benefits through health maintenance organizations (healthcare expenses were not submitted to the Centers for Medicare & Medicaid Services for payment, and therefore claims were not in the database or were incomplete); those with a prior claim history of <12 months (incomplete comorbidity history); and those who received a diagnosis of osteoporotic fracture (International Classification of Diseases, Ninth Revision, Clinical Modification [ICD-9-CM] code 733.1x) before the initial steroid injection.

We determined the incidence of osteoporotic wrist, hip, and spine fractures within 1, 2, and 8 years after LJSI, ESI, and TSI. Wrist, hip, and spine fractures were identified by ICD-9-CM diagnosis codes 733.12, 733.13, and 733.14, respectively. We also determined the number of steroid injections given before wrist, hip, or spine fracture or, if no fracture occurred, before death or the end of the data period.

Statistical Analysis

Multivariate Cox regression analysis was performed to evaluate the risk factors for wrist, spine, and hip fractures. The covariates in this model included age, sex, race, census region, Medicare buy-in status, Charlson Comorbidity Index (CCI),15 year, and number of steroid injections before fracture, death, or end of data period. Medicare buy-in status, which indicates whether the beneficiary received financial assistance in paying insurance premiums, was used as a proxy for socioeconomic status. CCI is used as a composite score of a patient’s general health status in terms of comorbidities.15,16 Four previously established categories17 were used to group CCIs in this study: 0 (none), 1 to 2 (low), 3 to 4 (moderate), and 5 or more (high). In addition, several diagnoses made within the 12 months before initial steroid injection were considered: osteoporosis (ICD-9-CM codes 733.0x, V82.81), Cushing syndrome (ICD-9-CM code 255.0), long-term (current) use of bisphosphonates (ICD-9-CM code V58.68), asymptomatic postmenopausal status (ICD-9-CM code V49.81), postmenopausal hormone replacement therapy (ICD-9-CM code V07.4), and long-term (current) use of steroids (ICD-9-CM code V58.65). The comparison of relative risk between any groups was reported as the adjusted hazard ratio (AHR), which is the ratio of the hazard rates of that particular outcome, taking into account inherent patient characteristics such as age, sex, and race as covariates. AHR of 1 corresponds to equivalent risk, AHR of >1 to elevated risk, and AHR of <1 to reduced risk.

Results

Using the 5% Medicare data for 2004 to 2011, we identified 275,999 Medicare beneficiaries who underwent LJSI, 93,943 who underwent ESI, and 32,311 who underwent TSI. During this period, TSI use increased, ESI use decreased, and LJSI use was relatively stable (Figure).

Figure.
LJSI patients underwent a mean (SD) of 3.0 (4.1) LJSIs; ESI patients, 3.2 (3.9) ESIs; and TSI patients, 7.5 (12.1) TSIs. Very few patients had more than 1 steroid injection site (Table 1).
Table 1.

The risk for osteoporotic spine fracture 1, 2, and 8 years after ESI, TSI, or LJSI was affected by age, race, sex, and CCI (P < .001 for all; Tables 2-4).

Table 2.
Patients with higher CCIs, females, older patients, and white patients had higher spine fracture risks. For ESI and LJSI patients, long-term steroid use was also associated with higher spine fracture risk at all examined time points.
Table 3.
For example, 8 years after ESI and LJSI, patients with long-term steroid use had 88% (P = .002) and 47% (P = .008) elevated risk of spine fracture, respectively. For TSI patients, spine fracture risk tended to be higher in patients with long-term steroid use 1 and 2 years after injection; this association was significant 8 years after injection (P = .046). ESI patients of lower socioeconomic status (Medicare buy-in) also had higher spine fracture risk at all examined time points, whereas LJSI and TSI patients did not. Two years after injection, each additional ESI decreased the relative risk of spine fracture by 2% (P < .001), each additional TSI decreased it by 1% (P = .009), and each additional LJSI decreased it by 4% (P < .001). 
Table 4.

The risk for osteoporotic hip fracture after 1 and 2 years was affected by age and number of LJSIs and TSIs but not by number of ESIs. Sex and CCI were also risk factors for hip fracture at 1 and 2 years for ESI and LJSI patients, as was race for LJSI patients. Risk for osteoporotic wrist fracture at 1 and 2 years was affected by sex and race for ESI and LJSI patients; age, race, CCI, and long-term steroid use were risk factors for TSI patients at all time points. Higher number of LJSIs, but not ESIs or TSIs, was associated with lower wrist fracture risk.

Discussion

ESIs continue to be used in the nonoperative treatment of low back pain, radicular leg pain, and spinal stenosis. Although the present study found ESI use increased in the Medicare population between 1994 and 2001,18 the trend is reversing, decreasing by 25%, with rates of 264 per 10,000 Medicare enrollees in 2004 and 194 per 10,000 enrollees in 2011. ESI use may have changed after systematic reviews revealed there was no clear evidence of the efficacy of ESIs in managing low back pain and radicular leg pain3,5,6 or spinal stenosis.4

Nevertheless, ESIs are widely used because of the perceived benefit balanced against the perceived rarity of adverse events.6 Even if patients recognize a low likelihood of significant benefit, they may accept ESI as preferable to surgery. In addition, most private payers require extensive nonoperative treatment before they will approve surgery as a treatment option.

In a study by Mandel and colleagues,14 ESI increased the risk of vertebral compression fractures by 21%, which in turn increased the risk of death.19 If accurate, these findings obviously would challenge the perception that ESI is a low-risk intervention. In contrast to the Mandel study,14 the present analysis of the Medicare population revealed no clinically relevant change in risk of osteoporotic spine fracture with each successive ESI after the initial injection. After the initial injection, each successive ESI decreased the relative risk of osteoporotic spine fracture by 2%, and each successive LJSI decreased it by 4%. Although statistically significant, the small change in relative risk may not be clinically relevant. However, taken cumulatively over a number of successive injections, these effects may be clinically relevant.

The data also showed that, after the initial injection, each successive ESI had no effect on risk of osteoporotic hip or wrist fracture, and each successive LJSI reduced the risk. Similar to earlier findings,20,21 long-term steroid use increased the risk of spine fracture in ESI and LJSI patients. Prolonged exposure to steroids may be necessary to reduce bone formation and increase bone breakdown.12

Although the study by Mandel and colleagues14 and our study both used administrative databases and survival analysis methods, conclusions differed. First, Mandel and colleagues14 used a study inclusion criterion of spine-related steroid injections, whereas we used a criterion of any steroid injection. Second, they used 50 years as the lower age for study inclusion, and we used 65 years. Third, to control for patients who had osteoporosis before study entry, they excluded those who had a fracture in an adjacent vertebra after kyphoplasty and vertebroplasty. It is unclear if patients who had osteoporotic fractures at other sites were excluded as well. Thus, the 2 cohorts may not be directly comparable.

Whereas Mandel and colleagues14 based their definition of osteoporotic spine fracture on a keyword search of a radiology database, we used a specific reportable ICD-9-CM diagnosis code. As a result, they may have overreported osteoporotic spine fractures, and we may have underreported. Finally, our sample was much larger than theirs. Given the relative rarity of osteoporotic fractures, a study with a larger sample may have more power to detect differences. In addition, unlike Mandel and colleagues,14 we focused on an injection cohort. We did not include or make comparisons with a no-injection cohort because our study hypothesis involved the potential systemic effects of steroid injections based on injection site. Although chronic steroid use was found to have a significant effect in our study, it is unclear to what extent the diagnosis code was used, during the comorbidity assessment or only in the event of steroid-related complications.

Our study also found that, after the initial injection, each successive LJSI decreased the risk of osteoporotic wrist fracture by 10%, and each successive TSI decreased the risk of osteoporotic hip fracture by 5%. It is plausible these injections allowed improved mobility, mitigating the effects of osteoporosis induced by inactivity and lack of resistance training. It is also possible that improved mobility limited falls.

In summary, this analysis of the Medicare claims database revealed that ESI, TSI, and LJSI decreased osteoporotic spine fracture risk. However, the effect was small and may not be clinically meaningful. After the initial injection, successive ESIs had no effect on the risk of osteoporotic hip or wrist fracture, and successive LJSIs reduced the risk of osteoporotic wrist fracture, perhaps because of improved mobility. Prolonged oral steroid use increased spine fracture risk in ESI and LJSI patients. More studies are needed to evaluate the risk-benefit profile of steroid injections.

References

1. Pethö G, Reeh PW. Sensory and signaling mechanisms of bradykinin, eicosanoids, platelet-activating factor, and nitric oxide in peripheral nociceptors. Physiol Rev. 2012;92(4):1699-1775.

2. Saal J. The role of inflammation in lumbar pain. Spine. 1995;20(16):1821-1827.

3. Choi HJ, Hahn S, Kim CH, et al. Epidural steroid injection therapy for low back pain: a meta-analysis. Int J Technol Assess Health Care. 2013;29(3):244-253.

4. Chou R, Loeser JD, Owens DK, et al; American Pain Society Low Back Pain Guideline Panel. Interventional therapies, surgery, and interdisciplinary rehabilitation for low back pain: an evidence-based clinical practice guideline from the American Pain Society. Spine. 2009;34(10):1066-1077. 

5. Savigny P, Watson P, Underwood M; Guideline Development Group. Early management of persistent non-specific low back pain: summary of NICE guidance. BMJ. 2009;338:b1805.

6. Staal JB, de Bie RA, de Vet HC, Hildebrandt J, Nelemans P. Injection therapy for subacute and chronic low back pain: an updated Cochrane review. Spine. 2009;34(1):49-59.

7. Angeli A, Guglielmi G, Dovio A, et al. High prevalence of asymptomatic vertebral fractures in post-menopausal women receiving chronic glucocorticoid therapy: a cross-sectional outpatient study. Bone. 2006;39(2):253-259.

8. Donnan PT, Libby G, Boyter AC, Thompson P. The population risk of fractures attributable to oral corticosteroids. Pharmacoepidemiol Drug Saf. 2005;14(3):177-186.

9. Dubois EF, Wagemans MF, Verdouw BC, et al. Lack of relationships between cumulative methylprednisolone dose and bone mineral density in healthy men and postmenopausal women with chronic low back pain. Clin Rheumatol. 2003;22(1):12-17.

10. Yi Y, Hwang B, Son H, Cheong I. Low bone mineral density, but not epidural steroid injection, is associated with fracture in postmenopausal women with low back pain. Pain Physician. 2012;15(6):441-449. 

11. Al-Shoha A, Rao DS, Schilling J, Peterson E, Mandel S. Effect of epidural steroid injection on bone mineral density and markers of bone turnover in postmenopausal women. Spine. 2012;37(25):E1567-E1571. 

12. Kang SS, Hwang BM, Son H, Cheong IY, Lee SJ, Chung TY. Changes in bone mineral density in postmenopausal women treated with epidural steroid injections for lower back pain. Pain Physician. 2012;15(3):229-236.

13. Kim S, Hwang B. Relationship between bone mineral density and the frequent administration of epidural steroid injections in postmenopausal women with low back pain. Pain Res Manag. 2014;19(1):30-34. 

14. Mandel S, Schilling J, Peterson E, Rao DS, Sanders W. A retrospective analysis of vertebral body fractures following epidural steroid injections. J Bone Joint Surg Am. 2013;95(11):961-964. 

15. Charlson ME, Pompei P, Ales KL, MacKenzie CR. A new method of classifying prognostic comorbidity in longitudinal studies: development and validation. J Chronic Dis. 1987;40(5):373-383.

16. Deyo RA, Cherkin DC, Ciol MA. Adapting a clinical comorbidity index for use with ICD-9-CM administrative databases. J Clin Epidemiol. 1992;45(6):613-619.

17. Murray SB, Bates DW, Ngo L, Ufberg JW, Shapiro NI. Charlson index is associated with one-year mortality in emergency department patients with suspected infection. Acad Emerg Med. 2006;13(5):530-536.

18. Friedly J, Chan L, Deyo R. Increases in lumbosacral injections in the Medicare population: 1994 to 2001. Spine. 2007;32(16):1754-1760.

19. Puisto V, Rissanen H, Heliövaara M, et al. Vertebral fracture and cause-specific mortality: a prospective population study of 3,210 men and 3,730 women with 30 years of follow-up. Eur Spine J. 2011;20(12):2181-2186. 

20. Lee YH, Woo JH, Choi SJ, Ji JD, Song GG. Effects of low-dose corticosteroids on the bone mineral density of patients with rheumatoid arthritis: a meta-analysis. J Investig Med. 2008;56(8):1011-1018.

21. Lukert BP, Raisz LG. Glucocorticoid-induced osteoporosis. Rheum Dis Clin North Am. 1994;20(3):629-650.

References

1. Pethö G, Reeh PW. Sensory and signaling mechanisms of bradykinin, eicosanoids, platelet-activating factor, and nitric oxide in peripheral nociceptors. Physiol Rev. 2012;92(4):1699-1775.

2. Saal J. The role of inflammation in lumbar pain. Spine. 1995;20(16):1821-1827.

3. Choi HJ, Hahn S, Kim CH, et al. Epidural steroid injection therapy for low back pain: a meta-analysis. Int J Technol Assess Health Care. 2013;29(3):244-253.

4. Chou R, Loeser JD, Owens DK, et al; American Pain Society Low Back Pain Guideline Panel. Interventional therapies, surgery, and interdisciplinary rehabilitation for low back pain: an evidence-based clinical practice guideline from the American Pain Society. Spine. 2009;34(10):1066-1077. 

5. Savigny P, Watson P, Underwood M; Guideline Development Group. Early management of persistent non-specific low back pain: summary of NICE guidance. BMJ. 2009;338:b1805.

6. Staal JB, de Bie RA, de Vet HC, Hildebrandt J, Nelemans P. Injection therapy for subacute and chronic low back pain: an updated Cochrane review. Spine. 2009;34(1):49-59.

7. Angeli A, Guglielmi G, Dovio A, et al. High prevalence of asymptomatic vertebral fractures in post-menopausal women receiving chronic glucocorticoid therapy: a cross-sectional outpatient study. Bone. 2006;39(2):253-259.

8. Donnan PT, Libby G, Boyter AC, Thompson P. The population risk of fractures attributable to oral corticosteroids. Pharmacoepidemiol Drug Saf. 2005;14(3):177-186.

9. Dubois EF, Wagemans MF, Verdouw BC, et al. Lack of relationships between cumulative methylprednisolone dose and bone mineral density in healthy men and postmenopausal women with chronic low back pain. Clin Rheumatol. 2003;22(1):12-17.

10. Yi Y, Hwang B, Son H, Cheong I. Low bone mineral density, but not epidural steroid injection, is associated with fracture in postmenopausal women with low back pain. Pain Physician. 2012;15(6):441-449. 

11. Al-Shoha A, Rao DS, Schilling J, Peterson E, Mandel S. Effect of epidural steroid injection on bone mineral density and markers of bone turnover in postmenopausal women. Spine. 2012;37(25):E1567-E1571. 

12. Kang SS, Hwang BM, Son H, Cheong IY, Lee SJ, Chung TY. Changes in bone mineral density in postmenopausal women treated with epidural steroid injections for lower back pain. Pain Physician. 2012;15(3):229-236.

13. Kim S, Hwang B. Relationship between bone mineral density and the frequent administration of epidural steroid injections in postmenopausal women with low back pain. Pain Res Manag. 2014;19(1):30-34. 

14. Mandel S, Schilling J, Peterson E, Rao DS, Sanders W. A retrospective analysis of vertebral body fractures following epidural steroid injections. J Bone Joint Surg Am. 2013;95(11):961-964. 

15. Charlson ME, Pompei P, Ales KL, MacKenzie CR. A new method of classifying prognostic comorbidity in longitudinal studies: development and validation. J Chronic Dis. 1987;40(5):373-383.

16. Deyo RA, Cherkin DC, Ciol MA. Adapting a clinical comorbidity index for use with ICD-9-CM administrative databases. J Clin Epidemiol. 1992;45(6):613-619.

17. Murray SB, Bates DW, Ngo L, Ufberg JW, Shapiro NI. Charlson index is associated with one-year mortality in emergency department patients with suspected infection. Acad Emerg Med. 2006;13(5):530-536.

18. Friedly J, Chan L, Deyo R. Increases in lumbosacral injections in the Medicare population: 1994 to 2001. Spine. 2007;32(16):1754-1760.

19. Puisto V, Rissanen H, Heliövaara M, et al. Vertebral fracture and cause-specific mortality: a prospective population study of 3,210 men and 3,730 women with 30 years of follow-up. Eur Spine J. 2011;20(12):2181-2186. 

20. Lee YH, Woo JH, Choi SJ, Ji JD, Song GG. Effects of low-dose corticosteroids on the bone mineral density of patients with rheumatoid arthritis: a meta-analysis. J Investig Med. 2008;56(8):1011-1018.

21. Lukert BP, Raisz LG. Glucocorticoid-induced osteoporosis. Rheum Dis Clin North Am. 1994;20(3):629-650.

Issue
The American Journal of Orthopedics - 46(5)
Issue
The American Journal of Orthopedics - 46(5)
Page Number
E293-E300
Page Number
E293-E300
Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Article PDF Media

Robot-assisted prostatectomy providing better outcomes

Article Type
Changed
Fri, 01/04/2019 - 13:41

 

Robot-assisted radical prostatectomy shows better early postoperative outcomes than does laparoscopic radical prostatectomy, but the differences between the two surgical approaches disappeared by the 6-month follow-up.

Dr. Hiroyuki Koike and his colleagues at Wakayama (Japan) Medical University Hospital conducted a study of two groups of patients treated for localized prostate cancer. One group of 229 patients underwent laparoscopic radical prostatectomy (LRP) between July 2007 and July 2013. The other group of 115 patients had robot-assisted radical prostatectomy (RARP) between December 2012 and August 2014 (J Robot Surg. 2017;11[3]:325-31).

Master Video/Shutterstock


The patients were given health-related quality of life self-assessment surveys prior to surgery and at 3, 6, and 12 months post surgery. In addition, a generic questionnaire, the eight-item Short-Form Health Survey, was used to assess a physical component summary (PCS) and a mental component summary (MCS). The Expanded Prostate Cancer Index of Prostate, which covers four domains – urinary, sexual, bowel, and hormonal – was used as a disease-specific measure, and the response rates for both LRP and RARP at each follow-up interval were over 80%.
 

 

“The RARP group showed significantly better scores in urinary summary and all urinary subscales at postoperative 3-month follow-up. However, these differences disappeared at postoperative 6 and 12-month follow-up,” the investigators wrote. For the urinary summary score, LRP significantly underperformed, compared with RARP, with scores of 63.3 vs. 75.8, respectively, after 3 months. In addition, the bowel function score was superior for RARP, compared with LRP, at 96.9 vs. 92.9, respectively. Sexual function results were similar, with RARP and LRP scores of 2.8 vs. 0.

The general measures of the PCS and MCS also favored RARP. At the 3-month follow-up, PCS (51.3 vs. 48.1) and MCS (50 vs. 47.8) scores were higher for RARP, compared with LRP.

“It is unclear why our superiority of urinary function in RARP was observed only in early period. However, we can speculate several reasons for better urinary function in RARP group. First, we were able to treat the apex area more delicately with RARP. Second, some of the new techniques which we employed after the introduction of RARP could influence the urinary continence recovery,” the investigators wrote.

The authors had no relevant financial disclosures.

Publications
Topics
Sections

 

Robot-assisted radical prostatectomy shows better early postoperative outcomes than does laparoscopic radical prostatectomy, but the differences between the two surgical approaches disappeared by the 6-month follow-up.

Dr. Hiroyuki Koike and his colleagues at Wakayama (Japan) Medical University Hospital conducted a study of two groups of patients treated for localized prostate cancer. One group of 229 patients underwent laparoscopic radical prostatectomy (LRP) between July 2007 and July 2013. The other group of 115 patients had robot-assisted radical prostatectomy (RARP) between December 2012 and August 2014 (J Robot Surg. 2017;11[3]:325-31).

Master Video/Shutterstock


The patients were given health-related quality of life self-assessment surveys prior to surgery and at 3, 6, and 12 months post surgery. In addition, a generic questionnaire, the eight-item Short-Form Health Survey, was used to assess a physical component summary (PCS) and a mental component summary (MCS). The Expanded Prostate Cancer Index of Prostate, which covers four domains – urinary, sexual, bowel, and hormonal – was used as a disease-specific measure, and the response rates for both LRP and RARP at each follow-up interval were over 80%.
 

 

“The RARP group showed significantly better scores in urinary summary and all urinary subscales at postoperative 3-month follow-up. However, these differences disappeared at postoperative 6 and 12-month follow-up,” the investigators wrote. For the urinary summary score, LRP significantly underperformed, compared with RARP, with scores of 63.3 vs. 75.8, respectively, after 3 months. In addition, the bowel function score was superior for RARP, compared with LRP, at 96.9 vs. 92.9, respectively. Sexual function results were similar, with RARP and LRP scores of 2.8 vs. 0.

The general measures of the PCS and MCS also favored RARP. At the 3-month follow-up, PCS (51.3 vs. 48.1) and MCS (50 vs. 47.8) scores were higher for RARP, compared with LRP.

“It is unclear why our superiority of urinary function in RARP was observed only in early period. However, we can speculate several reasons for better urinary function in RARP group. First, we were able to treat the apex area more delicately with RARP. Second, some of the new techniques which we employed after the introduction of RARP could influence the urinary continence recovery,” the investigators wrote.

The authors had no relevant financial disclosures.

 

Robot-assisted radical prostatectomy shows better early postoperative outcomes than does laparoscopic radical prostatectomy, but the differences between the two surgical approaches disappeared by the 6-month follow-up.

Dr. Hiroyuki Koike and his colleagues at Wakayama (Japan) Medical University Hospital conducted a study of two groups of patients treated for localized prostate cancer. One group of 229 patients underwent laparoscopic radical prostatectomy (LRP) between July 2007 and July 2013. The other group of 115 patients had robot-assisted radical prostatectomy (RARP) between December 2012 and August 2014 (J Robot Surg. 2017;11[3]:325-31).

Master Video/Shutterstock


The patients were given health-related quality of life self-assessment surveys prior to surgery and at 3, 6, and 12 months post surgery. In addition, a generic questionnaire, the eight-item Short-Form Health Survey, was used to assess a physical component summary (PCS) and a mental component summary (MCS). The Expanded Prostate Cancer Index of Prostate, which covers four domains – urinary, sexual, bowel, and hormonal – was used as a disease-specific measure, and the response rates for both LRP and RARP at each follow-up interval were over 80%.
 

 

“The RARP group showed significantly better scores in urinary summary and all urinary subscales at postoperative 3-month follow-up. However, these differences disappeared at postoperative 6 and 12-month follow-up,” the investigators wrote. For the urinary summary score, LRP significantly underperformed, compared with RARP, with scores of 63.3 vs. 75.8, respectively, after 3 months. In addition, the bowel function score was superior for RARP, compared with LRP, at 96.9 vs. 92.9, respectively. Sexual function results were similar, with RARP and LRP scores of 2.8 vs. 0.

The general measures of the PCS and MCS also favored RARP. At the 3-month follow-up, PCS (51.3 vs. 48.1) and MCS (50 vs. 47.8) scores were higher for RARP, compared with LRP.

“It is unclear why our superiority of urinary function in RARP was observed only in early period. However, we can speculate several reasons for better urinary function in RARP group. First, we were able to treat the apex area more delicately with RARP. Second, some of the new techniques which we employed after the introduction of RARP could influence the urinary continence recovery,” the investigators wrote.

The authors had no relevant financial disclosures.

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

FROM JOURNAL OF ROBOTIC SURGERY

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
CME ID
148188
Vitals

 

Key clinical point: Robotic-assisted radical prostatectomy provides better outcomes than laparoscopic procedures.

Major finding: Quality-of-life score for robotic-assisted radical prostatectomy was higher in all urinary categories after 3 months.

Data source: Postop survey results from patients with localized prostate cancer who underwent laparoscopic radical prostatectomy (n = 229) or robot-assisted radical prostatectomy (n = 115).

Disclosures: The investigators had no financial disclosures to report.

Disqus Comments
Default

FDA approves pembrolizumab for gastric and GEJ adenocarcinoma

Article Type
Changed
Wed, 05/26/2021 - 13:51

The Food and Drug Administration has approved pembrolizumab (Keytruda) for the treatment of patients with recurrent locally advanced or metastatic gastric or gastroesophageal junction (GEJ) adenocarcinoma, in cases where tests confirm that the tumors contain programmed death–ligand 1 and where the disease is progressing on or after two or more prior lines of therapy.

 

Pembrolizumab has been approved in the United States since 2014 for the treatment of melanoma, with subsequent approvals for treatment of non–small-cell lung cancer, head and neck squamous cell carcinoma, Hodgkin lymphoma, and several other advanced cancers.

Purple FDA logo.
The drug, now approved under the FDA’s accelerated approval regulations for the current indication in a 50-mg and 100-mg injection, blocks the interaction between the PD-1 protein and its ligands.

The approval comes on the basis of the nonrandomized, open label KEYNOTE-059 trial, which enrolled 259 patients with gastric or GEJ adenocarcinoma that progressed on at least two prior systemic treatments for advanced disease. Of the enrollees, 143 patients had tumors with a PD-L1 Combined Positive Score of 1 or greater. The primary trial outcome, the objective response rate for these 143 patients, was 13.3% (95% confidence interval; 8.2-20), with a complete response rate of 1.4% and a partial response rate of 11.9%. The duration of response ranged from at least 2.8 months to at least 19.4 months.

Continued approval for the new indication will depend upon further demonstration of a clinical benefit in confirmatory trials.

Publications
Topics
Sections

The Food and Drug Administration has approved pembrolizumab (Keytruda) for the treatment of patients with recurrent locally advanced or metastatic gastric or gastroesophageal junction (GEJ) adenocarcinoma, in cases where tests confirm that the tumors contain programmed death–ligand 1 and where the disease is progressing on or after two or more prior lines of therapy.

 

Pembrolizumab has been approved in the United States since 2014 for the treatment of melanoma, with subsequent approvals for treatment of non–small-cell lung cancer, head and neck squamous cell carcinoma, Hodgkin lymphoma, and several other advanced cancers.

Purple FDA logo.
The drug, now approved under the FDA’s accelerated approval regulations for the current indication in a 50-mg and 100-mg injection, blocks the interaction between the PD-1 protein and its ligands.

The approval comes on the basis of the nonrandomized, open label KEYNOTE-059 trial, which enrolled 259 patients with gastric or GEJ adenocarcinoma that progressed on at least two prior systemic treatments for advanced disease. Of the enrollees, 143 patients had tumors with a PD-L1 Combined Positive Score of 1 or greater. The primary trial outcome, the objective response rate for these 143 patients, was 13.3% (95% confidence interval; 8.2-20), with a complete response rate of 1.4% and a partial response rate of 11.9%. The duration of response ranged from at least 2.8 months to at least 19.4 months.

Continued approval for the new indication will depend upon further demonstration of a clinical benefit in confirmatory trials.

The Food and Drug Administration has approved pembrolizumab (Keytruda) for the treatment of patients with recurrent locally advanced or metastatic gastric or gastroesophageal junction (GEJ) adenocarcinoma, in cases where tests confirm that the tumors contain programmed death–ligand 1 and where the disease is progressing on or after two or more prior lines of therapy.

 

Pembrolizumab has been approved in the United States since 2014 for the treatment of melanoma, with subsequent approvals for treatment of non–small-cell lung cancer, head and neck squamous cell carcinoma, Hodgkin lymphoma, and several other advanced cancers.

Purple FDA logo.
The drug, now approved under the FDA’s accelerated approval regulations for the current indication in a 50-mg and 100-mg injection, blocks the interaction between the PD-1 protein and its ligands.

The approval comes on the basis of the nonrandomized, open label KEYNOTE-059 trial, which enrolled 259 patients with gastric or GEJ adenocarcinoma that progressed on at least two prior systemic treatments for advanced disease. Of the enrollees, 143 patients had tumors with a PD-L1 Combined Positive Score of 1 or greater. The primary trial outcome, the objective response rate for these 143 patients, was 13.3% (95% confidence interval; 8.2-20), with a complete response rate of 1.4% and a partial response rate of 11.9%. The duration of response ranged from at least 2.8 months to at least 19.4 months.

Continued approval for the new indication will depend upon further demonstration of a clinical benefit in confirmatory trials.

Publications
Publications
Topics
Article Type
Click for Credit Status
Ready
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default