Tumor Necrosis Factor Inhibitors May Reduce Cardiovascular Morbidity in Patients With Psoriasis

Article Type
Changed
Thu, 02/06/2020 - 12:07
Display Headline
Tumor Necrosis Factor Inhibitors May Reduce Cardiovascular Morbidity in Patients With Psoriasis

The connection between psoriasis and increased major adverse cardiovascular events (MACEs) has been well studied. 1,2 Although treatment of psoriasis can improve skin and joint symptoms, it is less clear whether therapies may mitigate the increased risk for cardiovascular comorbidities. Tumor necrosis factor (TNF) inhibitors in particular have been studied with great interest given the role of TNF in vascular and metabolic functions. 3 Using a retrospective cohort design, Wu and colleagues 4 examined if treatment with TNF inhibitors in patients with psoriasis would be associated with a lower risk for MACEs compared to phototherapy. Results suggested a significantly lower hazard of MACEs in patients using TNF inhibitors vs patients treated with phototherapy (adjusted hazard ratio, 0.77; P = .046). Moreover, based on these findings, they calculated that treating approximately 161 patients with TNF inhibitors rather than phototherapy would result in 1 less MACE per year overall. 4

Patients with psoriasis have been shown to have a greater noncalcified coronary plaque burden and prevalence of high-risk plaque compared to healthy patients.5 Lerman and colleagues5 measured the coronary plaque burden of 105 patients with psoriasis and 25 healthy volunteers using coronary computed tomography angiography. Although the patients were on average 10 years younger and had lower cardiovascular risk as measured by traditional risk scores, patients with psoriasis were found to have a greater noncalcified coronary plaque burden compared to 100 patients with hyperlipidemia. This burden was associated with an increased prevalence of high-risk plaques. Furthermore, in patients followed for 1 year, improvements in psoriasis severity were associated with reductions in noncalcified coronary plaque burden, though this finding was across all treatment modalities. However, there was no significant difference in calcified coronary plaque burden associated with reduced psoriasis severity.5

Moreover, Pina et al6 conducted a prospective study evaluating the use of the TNF inhibitor adalimumab to improve endothelial function and arterial stiffness in patients with moderate to severe psoriasis. Among 29 patients, they found a significant improvement in endothelial function as measured by flow-mediated dilatation after 6 months of adalimumab therapy, with a mean increase from 6.19% to 7.46% (P=.008). They also reported decreases in arterial stiffness by pulse wave velocity (P=.03). Despite a small sample size, these findings provide 2 potential mechanisms by which TNF inhibitor therapy may reduce the risk for cardiovascular events.6



A retrospective cohort study evaluating data from the Kaiser Permanente Southern California health plan assessed whether TNF inhibitor therapy was associated with a lower risk for MACE in patients with psoriasis.7 A total of 18,194 patients were included; of these, 1463 received TNF inhibitor therapy for at least 2 months. After controlling for other variables, including age at psoriasis diagnosis, sex, race/ethnicity, and other cardiovascular risk factors (eg, history of smoking or alcohol use; use of clopidogrel, antihypertensive agents, antihyperlipidemics, or anticoagulants), patients in the TNF inhibitor cohort demonstrated a significantly lower MACE hazard ratio compared to patients treated with topicals (hazard ratio, 0.80; 95% confidence interval, 0.66-0.98; P<.05).7

Conversely, a randomized, placebo-controlled trial of 107 patients found no difference in vascular inflammation of the ascending aorta and the carotids after 16 weeks of adalimumab treatment vs placebo. In this study, however, most patients had only moderate psoriasis based on a mean psoriasis area and severity index score of 9.8.8 Given studies finding higher risk burden in patients with more severe skin disease,2 it is possible that the effect of TNF inhibitor therapy may not be as pronounced in patients with less skin involvement. There was a significant effect on C-reactive protein levels in patients receiving TNF inhibitor therapy compared to placebo at 16 weeks (P=.012), suggesting TNF does play some role in systemic inflammation, and it is possible it may exert cardiovascular effects through a mechanism other than vascular inflammation.8

A second double-blind, randomized trial reported similar results.9 Among 97 patients randomized to receive adalimumab, placebo, or phototherapy, no significant difference in vascular inflammation was found after 12 weeks of therapy. In contrast, levels of C-reactive protein, IL-6, and glycoprotein acetylation were markedly reduced. The authors also reported adverse effects of adalimumab therapy on lipid metabolism with reduced cholesterol efflux capacity, a marker of ability of high-density-lipoprotein particles to perform reverse cholesterol transport, and high-density-lipoprotein particles, suggesting these effects may counteract some of the anti-inflammatory effects of TNF inhibitors.9



A growing body of data regarding the effect of TNF inhibitors on cardiovascular morbidity in patients with psoriasis is being collected, but no strong conclusions can be made. Given the disconnect of findings across these studies, it is possible that we have yet to elucidate the full mechanism by which TNF inhibitors may affect cardiovascular health. However, there may be additional confounding factors or patient characteristics at play. More large, prospective, randomized, controlled studies are needed to further understand this relationship.

References
  1. Ogdie A, Yu Y, Haynes K, et al. Risk of major cardiovascular events in patients with psoriatic arthritis, psoriasis and rheumatoid arthritis: a population-based cohort study. Ann Rheum Dis. 2015;74:326-332.
  2. Ahlehoff O, Gislason GH, Charlot M, et al. Psoriasis is associated with clinically significant cardiovascular risk: a Danish nationwide cohort study. J Intern Med. 2011;270:147-157.
  3. Kölliker Frers RA, Bisoendial RJ, Montoya SF, et al. Psoriasis and cardiovascular risk: immune-mediated crosstalk between metabolic, vascular, and autoimmune inflammation. Int J Cardiol Metab Endocr. 2015;6:43-54.
  4. Wu JJ, Sundaram M, Cloutier M, et al. The risk of cardiovascular events in psoriasis patients treated with tumor necrosis factor-α inhibitors versus phototherapy: an observational cohort study. J Am Acad Dermatol. 2018;79:60-68.
  5. Lerman JB, Joshi AA, Chaturvedi A, et al. Coronary plaque characterization in psoriasis reveals high-risk features that improve after treatment in a prospective observational study. Circulation. 2017;136:263-276.
  6. Pina T, Corrales A, Lopez-Mejias R, et al. Anti-tumor necrosis factor-α therapy improves endothelial function and arterial stiffness in patients with moderate to severe psoriasis: a 6 month prospective study. J Dermatol. 2016;43:1267-1272.
  7. Wu JJ, Joshi AA, Reddy SP, et al. Anti-inflammatory therapy with tumor necrosis factor inhibitors is associated with reduced risk of major adverse cardiovascular events in psoriasis [published online March 24, 2018]. J Eur Acad Dermatol Venereol. doi:10.1111/jdv.14951.
  8. Bissonnette R, Harel F, Krueger JG, et al. TNF-α antagonist and vascular inflammation patients with psoriasis vulgaris: a randomized placebo-controlled study. J Invest Dermatol. 2017;137:1638-1645 .
  9. Mehta NN, Shin DB, Joshi AA, et al. Effect of 2 psoriasis treatments on vascular inflammation and novel inflammatory cardiovascular biomarkers: a randomized placebo-controlled trial. Circ Cardiovasc Imaging. 2018;11:e007394.
Article PDF
Author and Disclosure Information

Dr. Lee is from the Department of Medicine, Santa Barbara Cottage Hospital, California. Dr. Amin is from the Department of Dermatology, Kaiser Permanente Los Angeles Medical Center, California. Dr. Wu is from Dermatology Research and Education Foundation, Irvine, California.

Drs. Lee and Amin report no conflict of interest. Dr. Wu is a consultant for AbbVie; Almirall; Amgen Inc; Bristol-Myers Squibb; Celgene Corporation; Dermira Inc; Dr. Reddy’s Laboratories; Eli Lilly and Company; Janssen Biotech, Inc; LEO Pharma; Novartis; Ortho Dermatologics; Promius Pharma; Regeneron Pharmaceuticals, Inc; Sun Pharmaceutical Industries, Ltd; and UCB. He also is an investigator for AbbVie Inc; Amgen Inc; Eli Lilly and Company, Janssen Biotech, Inc; and Novartis. He also is a speaker for Celgene Corporation; Novartis; Sun Pharmaceutical Industries, Ltd; and UCB.

Correspondence: Jashin J. Wu, MD (jashinwu@gmail.com).

Issue
Cutis - 105(2)
Publications
Topics
Page Number
60-61
Sections
Author and Disclosure Information

Dr. Lee is from the Department of Medicine, Santa Barbara Cottage Hospital, California. Dr. Amin is from the Department of Dermatology, Kaiser Permanente Los Angeles Medical Center, California. Dr. Wu is from Dermatology Research and Education Foundation, Irvine, California.

Drs. Lee and Amin report no conflict of interest. Dr. Wu is a consultant for AbbVie; Almirall; Amgen Inc; Bristol-Myers Squibb; Celgene Corporation; Dermira Inc; Dr. Reddy’s Laboratories; Eli Lilly and Company; Janssen Biotech, Inc; LEO Pharma; Novartis; Ortho Dermatologics; Promius Pharma; Regeneron Pharmaceuticals, Inc; Sun Pharmaceutical Industries, Ltd; and UCB. He also is an investigator for AbbVie Inc; Amgen Inc; Eli Lilly and Company, Janssen Biotech, Inc; and Novartis. He also is a speaker for Celgene Corporation; Novartis; Sun Pharmaceutical Industries, Ltd; and UCB.

Correspondence: Jashin J. Wu, MD (jashinwu@gmail.com).

Author and Disclosure Information

Dr. Lee is from the Department of Medicine, Santa Barbara Cottage Hospital, California. Dr. Amin is from the Department of Dermatology, Kaiser Permanente Los Angeles Medical Center, California. Dr. Wu is from Dermatology Research and Education Foundation, Irvine, California.

Drs. Lee and Amin report no conflict of interest. Dr. Wu is a consultant for AbbVie; Almirall; Amgen Inc; Bristol-Myers Squibb; Celgene Corporation; Dermira Inc; Dr. Reddy’s Laboratories; Eli Lilly and Company; Janssen Biotech, Inc; LEO Pharma; Novartis; Ortho Dermatologics; Promius Pharma; Regeneron Pharmaceuticals, Inc; Sun Pharmaceutical Industries, Ltd; and UCB. He also is an investigator for AbbVie Inc; Amgen Inc; Eli Lilly and Company, Janssen Biotech, Inc; and Novartis. He also is a speaker for Celgene Corporation; Novartis; Sun Pharmaceutical Industries, Ltd; and UCB.

Correspondence: Jashin J. Wu, MD (jashinwu@gmail.com).

Article PDF
Article PDF

The connection between psoriasis and increased major adverse cardiovascular events (MACEs) has been well studied. 1,2 Although treatment of psoriasis can improve skin and joint symptoms, it is less clear whether therapies may mitigate the increased risk for cardiovascular comorbidities. Tumor necrosis factor (TNF) inhibitors in particular have been studied with great interest given the role of TNF in vascular and metabolic functions. 3 Using a retrospective cohort design, Wu and colleagues 4 examined if treatment with TNF inhibitors in patients with psoriasis would be associated with a lower risk for MACEs compared to phototherapy. Results suggested a significantly lower hazard of MACEs in patients using TNF inhibitors vs patients treated with phototherapy (adjusted hazard ratio, 0.77; P = .046). Moreover, based on these findings, they calculated that treating approximately 161 patients with TNF inhibitors rather than phototherapy would result in 1 less MACE per year overall. 4

Patients with psoriasis have been shown to have a greater noncalcified coronary plaque burden and prevalence of high-risk plaque compared to healthy patients.5 Lerman and colleagues5 measured the coronary plaque burden of 105 patients with psoriasis and 25 healthy volunteers using coronary computed tomography angiography. Although the patients were on average 10 years younger and had lower cardiovascular risk as measured by traditional risk scores, patients with psoriasis were found to have a greater noncalcified coronary plaque burden compared to 100 patients with hyperlipidemia. This burden was associated with an increased prevalence of high-risk plaques. Furthermore, in patients followed for 1 year, improvements in psoriasis severity were associated with reductions in noncalcified coronary plaque burden, though this finding was across all treatment modalities. However, there was no significant difference in calcified coronary plaque burden associated with reduced psoriasis severity.5

Moreover, Pina et al6 conducted a prospective study evaluating the use of the TNF inhibitor adalimumab to improve endothelial function and arterial stiffness in patients with moderate to severe psoriasis. Among 29 patients, they found a significant improvement in endothelial function as measured by flow-mediated dilatation after 6 months of adalimumab therapy, with a mean increase from 6.19% to 7.46% (P=.008). They also reported decreases in arterial stiffness by pulse wave velocity (P=.03). Despite a small sample size, these findings provide 2 potential mechanisms by which TNF inhibitor therapy may reduce the risk for cardiovascular events.6



A retrospective cohort study evaluating data from the Kaiser Permanente Southern California health plan assessed whether TNF inhibitor therapy was associated with a lower risk for MACE in patients with psoriasis.7 A total of 18,194 patients were included; of these, 1463 received TNF inhibitor therapy for at least 2 months. After controlling for other variables, including age at psoriasis diagnosis, sex, race/ethnicity, and other cardiovascular risk factors (eg, history of smoking or alcohol use; use of clopidogrel, antihypertensive agents, antihyperlipidemics, or anticoagulants), patients in the TNF inhibitor cohort demonstrated a significantly lower MACE hazard ratio compared to patients treated with topicals (hazard ratio, 0.80; 95% confidence interval, 0.66-0.98; P<.05).7

Conversely, a randomized, placebo-controlled trial of 107 patients found no difference in vascular inflammation of the ascending aorta and the carotids after 16 weeks of adalimumab treatment vs placebo. In this study, however, most patients had only moderate psoriasis based on a mean psoriasis area and severity index score of 9.8.8 Given studies finding higher risk burden in patients with more severe skin disease,2 it is possible that the effect of TNF inhibitor therapy may not be as pronounced in patients with less skin involvement. There was a significant effect on C-reactive protein levels in patients receiving TNF inhibitor therapy compared to placebo at 16 weeks (P=.012), suggesting TNF does play some role in systemic inflammation, and it is possible it may exert cardiovascular effects through a mechanism other than vascular inflammation.8

A second double-blind, randomized trial reported similar results.9 Among 97 patients randomized to receive adalimumab, placebo, or phototherapy, no significant difference in vascular inflammation was found after 12 weeks of therapy. In contrast, levels of C-reactive protein, IL-6, and glycoprotein acetylation were markedly reduced. The authors also reported adverse effects of adalimumab therapy on lipid metabolism with reduced cholesterol efflux capacity, a marker of ability of high-density-lipoprotein particles to perform reverse cholesterol transport, and high-density-lipoprotein particles, suggesting these effects may counteract some of the anti-inflammatory effects of TNF inhibitors.9



A growing body of data regarding the effect of TNF inhibitors on cardiovascular morbidity in patients with psoriasis is being collected, but no strong conclusions can be made. Given the disconnect of findings across these studies, it is possible that we have yet to elucidate the full mechanism by which TNF inhibitors may affect cardiovascular health. However, there may be additional confounding factors or patient characteristics at play. More large, prospective, randomized, controlled studies are needed to further understand this relationship.

The connection between psoriasis and increased major adverse cardiovascular events (MACEs) has been well studied. 1,2 Although treatment of psoriasis can improve skin and joint symptoms, it is less clear whether therapies may mitigate the increased risk for cardiovascular comorbidities. Tumor necrosis factor (TNF) inhibitors in particular have been studied with great interest given the role of TNF in vascular and metabolic functions. 3 Using a retrospective cohort design, Wu and colleagues 4 examined if treatment with TNF inhibitors in patients with psoriasis would be associated with a lower risk for MACEs compared to phototherapy. Results suggested a significantly lower hazard of MACEs in patients using TNF inhibitors vs patients treated with phototherapy (adjusted hazard ratio, 0.77; P = .046). Moreover, based on these findings, they calculated that treating approximately 161 patients with TNF inhibitors rather than phototherapy would result in 1 less MACE per year overall. 4

Patients with psoriasis have been shown to have a greater noncalcified coronary plaque burden and prevalence of high-risk plaque compared to healthy patients.5 Lerman and colleagues5 measured the coronary plaque burden of 105 patients with psoriasis and 25 healthy volunteers using coronary computed tomography angiography. Although the patients were on average 10 years younger and had lower cardiovascular risk as measured by traditional risk scores, patients with psoriasis were found to have a greater noncalcified coronary plaque burden compared to 100 patients with hyperlipidemia. This burden was associated with an increased prevalence of high-risk plaques. Furthermore, in patients followed for 1 year, improvements in psoriasis severity were associated with reductions in noncalcified coronary plaque burden, though this finding was across all treatment modalities. However, there was no significant difference in calcified coronary plaque burden associated with reduced psoriasis severity.5

Moreover, Pina et al6 conducted a prospective study evaluating the use of the TNF inhibitor adalimumab to improve endothelial function and arterial stiffness in patients with moderate to severe psoriasis. Among 29 patients, they found a significant improvement in endothelial function as measured by flow-mediated dilatation after 6 months of adalimumab therapy, with a mean increase from 6.19% to 7.46% (P=.008). They also reported decreases in arterial stiffness by pulse wave velocity (P=.03). Despite a small sample size, these findings provide 2 potential mechanisms by which TNF inhibitor therapy may reduce the risk for cardiovascular events.6



A retrospective cohort study evaluating data from the Kaiser Permanente Southern California health plan assessed whether TNF inhibitor therapy was associated with a lower risk for MACE in patients with psoriasis.7 A total of 18,194 patients were included; of these, 1463 received TNF inhibitor therapy for at least 2 months. After controlling for other variables, including age at psoriasis diagnosis, sex, race/ethnicity, and other cardiovascular risk factors (eg, history of smoking or alcohol use; use of clopidogrel, antihypertensive agents, antihyperlipidemics, or anticoagulants), patients in the TNF inhibitor cohort demonstrated a significantly lower MACE hazard ratio compared to patients treated with topicals (hazard ratio, 0.80; 95% confidence interval, 0.66-0.98; P<.05).7

Conversely, a randomized, placebo-controlled trial of 107 patients found no difference in vascular inflammation of the ascending aorta and the carotids after 16 weeks of adalimumab treatment vs placebo. In this study, however, most patients had only moderate psoriasis based on a mean psoriasis area and severity index score of 9.8.8 Given studies finding higher risk burden in patients with more severe skin disease,2 it is possible that the effect of TNF inhibitor therapy may not be as pronounced in patients with less skin involvement. There was a significant effect on C-reactive protein levels in patients receiving TNF inhibitor therapy compared to placebo at 16 weeks (P=.012), suggesting TNF does play some role in systemic inflammation, and it is possible it may exert cardiovascular effects through a mechanism other than vascular inflammation.8

A second double-blind, randomized trial reported similar results.9 Among 97 patients randomized to receive adalimumab, placebo, or phototherapy, no significant difference in vascular inflammation was found after 12 weeks of therapy. In contrast, levels of C-reactive protein, IL-6, and glycoprotein acetylation were markedly reduced. The authors also reported adverse effects of adalimumab therapy on lipid metabolism with reduced cholesterol efflux capacity, a marker of ability of high-density-lipoprotein particles to perform reverse cholesterol transport, and high-density-lipoprotein particles, suggesting these effects may counteract some of the anti-inflammatory effects of TNF inhibitors.9



A growing body of data regarding the effect of TNF inhibitors on cardiovascular morbidity in patients with psoriasis is being collected, but no strong conclusions can be made. Given the disconnect of findings across these studies, it is possible that we have yet to elucidate the full mechanism by which TNF inhibitors may affect cardiovascular health. However, there may be additional confounding factors or patient characteristics at play. More large, prospective, randomized, controlled studies are needed to further understand this relationship.

References
  1. Ogdie A, Yu Y, Haynes K, et al. Risk of major cardiovascular events in patients with psoriatic arthritis, psoriasis and rheumatoid arthritis: a population-based cohort study. Ann Rheum Dis. 2015;74:326-332.
  2. Ahlehoff O, Gislason GH, Charlot M, et al. Psoriasis is associated with clinically significant cardiovascular risk: a Danish nationwide cohort study. J Intern Med. 2011;270:147-157.
  3. Kölliker Frers RA, Bisoendial RJ, Montoya SF, et al. Psoriasis and cardiovascular risk: immune-mediated crosstalk between metabolic, vascular, and autoimmune inflammation. Int J Cardiol Metab Endocr. 2015;6:43-54.
  4. Wu JJ, Sundaram M, Cloutier M, et al. The risk of cardiovascular events in psoriasis patients treated with tumor necrosis factor-α inhibitors versus phototherapy: an observational cohort study. J Am Acad Dermatol. 2018;79:60-68.
  5. Lerman JB, Joshi AA, Chaturvedi A, et al. Coronary plaque characterization in psoriasis reveals high-risk features that improve after treatment in a prospective observational study. Circulation. 2017;136:263-276.
  6. Pina T, Corrales A, Lopez-Mejias R, et al. Anti-tumor necrosis factor-α therapy improves endothelial function and arterial stiffness in patients with moderate to severe psoriasis: a 6 month prospective study. J Dermatol. 2016;43:1267-1272.
  7. Wu JJ, Joshi AA, Reddy SP, et al. Anti-inflammatory therapy with tumor necrosis factor inhibitors is associated with reduced risk of major adverse cardiovascular events in psoriasis [published online March 24, 2018]. J Eur Acad Dermatol Venereol. doi:10.1111/jdv.14951.
  8. Bissonnette R, Harel F, Krueger JG, et al. TNF-α antagonist and vascular inflammation patients with psoriasis vulgaris: a randomized placebo-controlled study. J Invest Dermatol. 2017;137:1638-1645 .
  9. Mehta NN, Shin DB, Joshi AA, et al. Effect of 2 psoriasis treatments on vascular inflammation and novel inflammatory cardiovascular biomarkers: a randomized placebo-controlled trial. Circ Cardiovasc Imaging. 2018;11:e007394.
References
  1. Ogdie A, Yu Y, Haynes K, et al. Risk of major cardiovascular events in patients with psoriatic arthritis, psoriasis and rheumatoid arthritis: a population-based cohort study. Ann Rheum Dis. 2015;74:326-332.
  2. Ahlehoff O, Gislason GH, Charlot M, et al. Psoriasis is associated with clinically significant cardiovascular risk: a Danish nationwide cohort study. J Intern Med. 2011;270:147-157.
  3. Kölliker Frers RA, Bisoendial RJ, Montoya SF, et al. Psoriasis and cardiovascular risk: immune-mediated crosstalk between metabolic, vascular, and autoimmune inflammation. Int J Cardiol Metab Endocr. 2015;6:43-54.
  4. Wu JJ, Sundaram M, Cloutier M, et al. The risk of cardiovascular events in psoriasis patients treated with tumor necrosis factor-α inhibitors versus phototherapy: an observational cohort study. J Am Acad Dermatol. 2018;79:60-68.
  5. Lerman JB, Joshi AA, Chaturvedi A, et al. Coronary plaque characterization in psoriasis reveals high-risk features that improve after treatment in a prospective observational study. Circulation. 2017;136:263-276.
  6. Pina T, Corrales A, Lopez-Mejias R, et al. Anti-tumor necrosis factor-α therapy improves endothelial function and arterial stiffness in patients with moderate to severe psoriasis: a 6 month prospective study. J Dermatol. 2016;43:1267-1272.
  7. Wu JJ, Joshi AA, Reddy SP, et al. Anti-inflammatory therapy with tumor necrosis factor inhibitors is associated with reduced risk of major adverse cardiovascular events in psoriasis [published online March 24, 2018]. J Eur Acad Dermatol Venereol. doi:10.1111/jdv.14951.
  8. Bissonnette R, Harel F, Krueger JG, et al. TNF-α antagonist and vascular inflammation patients with psoriasis vulgaris: a randomized placebo-controlled study. J Invest Dermatol. 2017;137:1638-1645 .
  9. Mehta NN, Shin DB, Joshi AA, et al. Effect of 2 psoriasis treatments on vascular inflammation and novel inflammatory cardiovascular biomarkers: a randomized placebo-controlled trial. Circ Cardiovasc Imaging. 2018;11:e007394.
Issue
Cutis - 105(2)
Issue
Cutis - 105(2)
Page Number
60-61
Page Number
60-61
Publications
Publications
Topics
Article Type
Display Headline
Tumor Necrosis Factor Inhibitors May Reduce Cardiovascular Morbidity in Patients With Psoriasis
Display Headline
Tumor Necrosis Factor Inhibitors May Reduce Cardiovascular Morbidity in Patients With Psoriasis
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Article PDF Media

Chronic cough in COPD linked to more severe disease

Article Type
Changed
Mon, 02/10/2020 - 09:26

In patients with chronic obstructive pulmonary disease (COPD), comorbid chronic cough is associated with more respiratory symptoms and health care utilization, decreased lung function, and increased inflammatory markers in blood, according to research published in CHEST.

The results indicate “that chronic cough in individuals with COPD is associated with a more severe disease phenotype, which could be helpful for stratifying management of COPD in the future,” wrote Eskild Landt, PhD, a research assistant at Zealand University Hospital in Køge, Denmark, and colleagues.

A study by published in the Journal of Allergy and Clinical Immunology: In Practice (2019;7[6]:1783-92.e8) indicated that in patients with asthma, chronic cough was associated with worse respiratory symptoms, more health care utilization, decreased lung function, and increased inflammatory markers in blood. Dr. Landt and colleagues hypothesized that patients with COPD and chronic cough had a similar pattern of disease severity.

To test their hypothesis, they identified individuals with COPD and chronic cough among 43,271 participants in the Copenhagen General Population Study, a population-based cohort study. The researchers defined COPD as a ratio of forced expiratory volume in 1 second (FEV1) to forced vital capacity (FVC) of less than 0.70 in individuals without asthma. Consecutive individuals answered questions about chronic cough, which was defined as a cough lasting more than 8 weeks, and responded to the Leicester Cough Questionnaire. They also underwent a physical health examination, including prebronchodilatory spirometry, and gave blood for biochemical analyses. The blood was analyzed for high-sensitive C-reactive protein, fibrinogen, leukocytes, eosinophils, neutrophils, and immunoglobulin E (i.e., inflammatory biomarkers).

Dr. Landt and colleagues identified 8,181 patients (19% of the population) with COPD, 796 (10%) of whom had chronic cough. Of the 33,364 participants without COPD, 1,585 (5%) had chronic cough. For patients with COPD and chronic cough, median total Leicester Cough Questionnaire score was 17.7, corresponding to 5.9 for the physical domain, 5.6 for the psychological domain, and 6.3 for the social domain.

Among participants with COPD, those with chronic cough had higher rates of sputum production (60% versus 8%), wheezing (46% versus 14%), dyspnea (66% versus 38%), chest pain or tightness (9% versus 4%), nighttime dyspnea (8% versus 3%), episodes of acute bronchitis or pneumonias in the past 10 years (45% versus 25%), and general practitioner visits in the past 12 months (53% versus 37%). In addition, these participants had lower FEV1% of predicted (81% versus 89%), lower ratio of FEV1 to FVC (0.64 versus 0.66), and higher levels of high-sensitive C-reactive protein, fibrinogen, leukocytes, neutrophils, eosinophils, and immunoglobulin E in blood.

“To our knowledge, this is the first study reporting Leicester Cough Questionnaire score for randomly selected individuals with COPD from a general population setting,” wrote Dr. Landt and colleagues. The study’s strengths include its randomly chosen population-based sample and investigator blinding to disease status and clinical outcome, they added. Some patients with the most severe types of COPD and chronic cough may not have attended the physical examination and participated in the study, however, and this factor could have biased the results. Furthermore, nearly the entire sample was white, so the results may not be generalizable to other ethnicities. “That said, we are not aware of results to suggest that our findings should not be relevant to individuals of all races,” wrote the investigators.

The study was funded by the private Lundbeck Foundation, as well as by the Danish Lung Association and the Danish Cancer Society. Several authors reported receiving grants and fees from AstraZeneca, GlaxoSmithKline, and Novartis that were unrelated to the study.

SOURCE: Landt E et al. CHEST. 2020 Jan 24. doi: 10.1016/j.chest.2019.12.038.

Publications
Topics
Sections

In patients with chronic obstructive pulmonary disease (COPD), comorbid chronic cough is associated with more respiratory symptoms and health care utilization, decreased lung function, and increased inflammatory markers in blood, according to research published in CHEST.

The results indicate “that chronic cough in individuals with COPD is associated with a more severe disease phenotype, which could be helpful for stratifying management of COPD in the future,” wrote Eskild Landt, PhD, a research assistant at Zealand University Hospital in Køge, Denmark, and colleagues.

A study by published in the Journal of Allergy and Clinical Immunology: In Practice (2019;7[6]:1783-92.e8) indicated that in patients with asthma, chronic cough was associated with worse respiratory symptoms, more health care utilization, decreased lung function, and increased inflammatory markers in blood. Dr. Landt and colleagues hypothesized that patients with COPD and chronic cough had a similar pattern of disease severity.

To test their hypothesis, they identified individuals with COPD and chronic cough among 43,271 participants in the Copenhagen General Population Study, a population-based cohort study. The researchers defined COPD as a ratio of forced expiratory volume in 1 second (FEV1) to forced vital capacity (FVC) of less than 0.70 in individuals without asthma. Consecutive individuals answered questions about chronic cough, which was defined as a cough lasting more than 8 weeks, and responded to the Leicester Cough Questionnaire. They also underwent a physical health examination, including prebronchodilatory spirometry, and gave blood for biochemical analyses. The blood was analyzed for high-sensitive C-reactive protein, fibrinogen, leukocytes, eosinophils, neutrophils, and immunoglobulin E (i.e., inflammatory biomarkers).

Dr. Landt and colleagues identified 8,181 patients (19% of the population) with COPD, 796 (10%) of whom had chronic cough. Of the 33,364 participants without COPD, 1,585 (5%) had chronic cough. For patients with COPD and chronic cough, median total Leicester Cough Questionnaire score was 17.7, corresponding to 5.9 for the physical domain, 5.6 for the psychological domain, and 6.3 for the social domain.

Among participants with COPD, those with chronic cough had higher rates of sputum production (60% versus 8%), wheezing (46% versus 14%), dyspnea (66% versus 38%), chest pain or tightness (9% versus 4%), nighttime dyspnea (8% versus 3%), episodes of acute bronchitis or pneumonias in the past 10 years (45% versus 25%), and general practitioner visits in the past 12 months (53% versus 37%). In addition, these participants had lower FEV1% of predicted (81% versus 89%), lower ratio of FEV1 to FVC (0.64 versus 0.66), and higher levels of high-sensitive C-reactive protein, fibrinogen, leukocytes, neutrophils, eosinophils, and immunoglobulin E in blood.

“To our knowledge, this is the first study reporting Leicester Cough Questionnaire score for randomly selected individuals with COPD from a general population setting,” wrote Dr. Landt and colleagues. The study’s strengths include its randomly chosen population-based sample and investigator blinding to disease status and clinical outcome, they added. Some patients with the most severe types of COPD and chronic cough may not have attended the physical examination and participated in the study, however, and this factor could have biased the results. Furthermore, nearly the entire sample was white, so the results may not be generalizable to other ethnicities. “That said, we are not aware of results to suggest that our findings should not be relevant to individuals of all races,” wrote the investigators.

The study was funded by the private Lundbeck Foundation, as well as by the Danish Lung Association and the Danish Cancer Society. Several authors reported receiving grants and fees from AstraZeneca, GlaxoSmithKline, and Novartis that were unrelated to the study.

SOURCE: Landt E et al. CHEST. 2020 Jan 24. doi: 10.1016/j.chest.2019.12.038.

In patients with chronic obstructive pulmonary disease (COPD), comorbid chronic cough is associated with more respiratory symptoms and health care utilization, decreased lung function, and increased inflammatory markers in blood, according to research published in CHEST.

The results indicate “that chronic cough in individuals with COPD is associated with a more severe disease phenotype, which could be helpful for stratifying management of COPD in the future,” wrote Eskild Landt, PhD, a research assistant at Zealand University Hospital in Køge, Denmark, and colleagues.

A study by published in the Journal of Allergy and Clinical Immunology: In Practice (2019;7[6]:1783-92.e8) indicated that in patients with asthma, chronic cough was associated with worse respiratory symptoms, more health care utilization, decreased lung function, and increased inflammatory markers in blood. Dr. Landt and colleagues hypothesized that patients with COPD and chronic cough had a similar pattern of disease severity.

To test their hypothesis, they identified individuals with COPD and chronic cough among 43,271 participants in the Copenhagen General Population Study, a population-based cohort study. The researchers defined COPD as a ratio of forced expiratory volume in 1 second (FEV1) to forced vital capacity (FVC) of less than 0.70 in individuals without asthma. Consecutive individuals answered questions about chronic cough, which was defined as a cough lasting more than 8 weeks, and responded to the Leicester Cough Questionnaire. They also underwent a physical health examination, including prebronchodilatory spirometry, and gave blood for biochemical analyses. The blood was analyzed for high-sensitive C-reactive protein, fibrinogen, leukocytes, eosinophils, neutrophils, and immunoglobulin E (i.e., inflammatory biomarkers).

Dr. Landt and colleagues identified 8,181 patients (19% of the population) with COPD, 796 (10%) of whom had chronic cough. Of the 33,364 participants without COPD, 1,585 (5%) had chronic cough. For patients with COPD and chronic cough, median total Leicester Cough Questionnaire score was 17.7, corresponding to 5.9 for the physical domain, 5.6 for the psychological domain, and 6.3 for the social domain.

Among participants with COPD, those with chronic cough had higher rates of sputum production (60% versus 8%), wheezing (46% versus 14%), dyspnea (66% versus 38%), chest pain or tightness (9% versus 4%), nighttime dyspnea (8% versus 3%), episodes of acute bronchitis or pneumonias in the past 10 years (45% versus 25%), and general practitioner visits in the past 12 months (53% versus 37%). In addition, these participants had lower FEV1% of predicted (81% versus 89%), lower ratio of FEV1 to FVC (0.64 versus 0.66), and higher levels of high-sensitive C-reactive protein, fibrinogen, leukocytes, neutrophils, eosinophils, and immunoglobulin E in blood.

“To our knowledge, this is the first study reporting Leicester Cough Questionnaire score for randomly selected individuals with COPD from a general population setting,” wrote Dr. Landt and colleagues. The study’s strengths include its randomly chosen population-based sample and investigator blinding to disease status and clinical outcome, they added. Some patients with the most severe types of COPD and chronic cough may not have attended the physical examination and participated in the study, however, and this factor could have biased the results. Furthermore, nearly the entire sample was white, so the results may not be generalizable to other ethnicities. “That said, we are not aware of results to suggest that our findings should not be relevant to individuals of all races,” wrote the investigators.

The study was funded by the private Lundbeck Foundation, as well as by the Danish Lung Association and the Danish Cancer Society. Several authors reported receiving grants and fees from AstraZeneca, GlaxoSmithKline, and Novartis that were unrelated to the study.

SOURCE: Landt E et al. CHEST. 2020 Jan 24. doi: 10.1016/j.chest.2019.12.038.

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM CHEST

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.

Documentation matters

Article Type
Changed
Tue, 02/04/2020 - 14:44

Quality over quantity

Documentation has always been part of a physician’s job. Historically, in the days of paper records, physicians saw a patient on rounds and immediately following, while still on the unit, wrote a daily note detailing the events, test results, and plans since the last note. Addenda were written over the course of the day and night as needed.

Dr. Erica Remer, on the board of directors of the American College of Physician Advisors and the advisory board of the Association of Clinical Documentation Improvement Specialists
Dr. Erica Remer

The medical record was a chronological itemization of the encounter. The chart told the patient’s story, hopefully legibly and without excessive rehashing of previous material. The discharge summary then encapsulated the hospitalization in several coherent paragraphs.

In the current electronic records environment, we are inundated with excessive and repetitious information, data without interpretation, differentials without diagnoses. Prepopulation of templated notes, defaults without edit, and dictation without revision have degraded our documentation to the point of unintelligibility. The chronological storytelling and trustworthiness of the medical record has become suspect.

The Centers for Medicare & Medicaid Services is touting its “Patients over Paperwork” initiative. The solution is flawed (that is, future relaxation of documentation requirements for professional billing) because the premise is delusive. Documentation isn’t fundamentally the problem. Having clinicians jump through regulatory hoops which do not advance patients’ care, and providers misunderstanding the requirements for level-of-service billing are the essential issues. Getting no training on how to properly document in medical school/residency and receiving no formative feedback on documentation throughout one’s career compounds the problem. Having clinical documentation serve too many masters, including compliance, quality, medicolegal, utilization review, and reimbursement, is also to blame. The advent of the electronic medical record was just the straw that broke the camel’s back.

Many hospitals now have a clinical documentation integrity (CDI) team which is tasked with querying the provider when the health record documentation is conflicting, imprecise, incomplete, illegible, ambiguous, or inconsistent. They are charged with getting practitioners to associate clinical indicators with diagnoses and to consider removal of diagnoses which do not seem clinically valid from the existing documentation. From this explanation, you might well conclude that the CDI specialist could generate a query on every patient if they were so inclined, and you would be correct. But the goal isn’t to torture the physician – it is to ensure that the medical record is accurately depicting the encounter.

You are not being asked for more documentation by the CDI team; they are entreating you for higher-quality documentation. Let me give you some pointers to ward off queries.

  • Tell the story. The most important goal of documentation is to clinically communicate to other caregivers. Think to yourself: “What would a fellow clinician need to know about this patient to understand why I drew those conclusions or to pick up where I left off?” At 2 a.m., that information, or lack thereof, could literally be a matter of life or death.
  • Tell the truth. Embellishing the record or including invalid diagnoses with the intent to increase the severity of illness resulting in a more favorable diagnosis-related group – the inpatient risk-adjustment system – is considered fraud.
  • You may like the convenience of copy forward, but do you relish reading other people’s copy and paste? Consider doing a documentation time-out. Before you copy and paste yesterday’s assessment and plan, stop and think: “Why is the patient still here? Why are we doing what we are doing?” If you choose to copy and paste, be certain to do mindful editing so the documentation represents the current situation and avoids redundancy. Appropriately editing copy and pasted documentation may prove more time consuming than generating a note de novo.
  • Translate findings into diagnoses using your best medical judgment. One man’s hypotension may be another health care provider’s shock. Coders are not clinical and are not permitted to make inferences. A potassium of 6.7 may be hyperkalemia or it may be spurious – only a clinician may make that determination using their clinical expertise and experience. The coder is not allowed to read your mind. You must explicitly draw the conclusion that a febrile patient with bacteremia, encephalopathy, hypoxemia, and a blood pressure of 85/60 is in septic shock.
  • Uncertain diagnoses (heralded by words such as: likely, possible, probable, suspected, rule out, etc.) which are not ruled out prior to discharge or demise are coded as if they were definitively present, for the inpatient technical side of hospital billing. This is distinctly different than the professional fee where you can only code definitive diagnoses. If you have a strong suspicion (not wild speculation) that a condition is present, best practice is to offer an uncertain diagnosis. Associate signs and symptoms with your most likely diagnosis: “Shortness of breath, pleuritic chest pain, and hypoxemia in the setting of cancer, probable pulmonary embolism.”
  • Evolve, resolve, remove, and recap. If an uncertain diagnosis is ruled in, take away the uncertainty. If it is ruled out, don’t have 4 days of copy and pasted: “Possible eosinophilic pneumonia.” You do not have to maintain a resolved diagnosis ad infinitum. It can drop off the diagnosis list but be sure to have it reappear in the discharge summary.
  • I know it can be a hASSLe to do excellent documentation, but it is critical for many reasons, most importantly for superlative patient care. More accurate coding and billing is an intended consequence. A: Acuity; S: Severity; S: Specificity (may affect the coding and the risk-adjustment implications. Acute systolic heart failure does not equal heart failure; type 2 diabetes mellitus with diabetic chronic kidney disease, stage 4 does not equal chronic kidney disease); and L: Linkage (of diagnosis with underlying cause or manifestation [e.g., because of, associated with, as a result of, secondary to, or from diabetic nephropathy, hypertensive encephalopathy]).
  • If you have the capability to keep a running summary throughout the hospital stay, do so and keep it updated. A few moments of daily careful editing and composing can save time and effort at the back end creating the discharge summary. The follow-up care provider can reconstruct the hospital course and it is your last chance to spin the narrative for the lawyers.
  • Read your documentation over. Ensure that it is clear, accurate, concise, and tells the story and the plans for the patient. Make sure that someone reading the note will know what you were thinking.
  • Set up a program to self-audit documentation where monthly or quarterly, you and your partners mutually review a certain number of records and give each other feedback. Design an assessment tool which rates the quality of documentation elements which your hospital/network/service line values (clarity, copy and paste, complete and specific diagnoses, etc.). You know who the best documenters are. Why do you think their documentation is superior? How can you emulate them?

Finally, answer CDI queries. The CDI specialist is your ally, not your enemy. They want you to get credit for taking care of sick and complex patients. They are not permitted to lead the provider, so don’t ask them what they want you to write. But, if you don’t understand the query or issue, have a conversation and get it clarified. It is in everyone’s best interest to get this right.

Documentation improves patient care and demonstrates that you provided excellent patient care. Put mentation back into documentation.

Dr. Remer was a practicing emergency physician for 25 years and a physician advisor for 4 years. She is on the board of directors of the American College of Physician Advisors and the advisory board of the Association of Clinical Documentation Improvement Specialists. She currently provides consulting services for provider education on documentation, CDI, and ICD-10 coding. Dr. Remer can be reached at eremer@icd10md.com

Publications
Topics
Sections

Quality over quantity

Quality over quantity

Documentation has always been part of a physician’s job. Historically, in the days of paper records, physicians saw a patient on rounds and immediately following, while still on the unit, wrote a daily note detailing the events, test results, and plans since the last note. Addenda were written over the course of the day and night as needed.

Dr. Erica Remer, on the board of directors of the American College of Physician Advisors and the advisory board of the Association of Clinical Documentation Improvement Specialists
Dr. Erica Remer

The medical record was a chronological itemization of the encounter. The chart told the patient’s story, hopefully legibly and without excessive rehashing of previous material. The discharge summary then encapsulated the hospitalization in several coherent paragraphs.

In the current electronic records environment, we are inundated with excessive and repetitious information, data without interpretation, differentials without diagnoses. Prepopulation of templated notes, defaults without edit, and dictation without revision have degraded our documentation to the point of unintelligibility. The chronological storytelling and trustworthiness of the medical record has become suspect.

The Centers for Medicare & Medicaid Services is touting its “Patients over Paperwork” initiative. The solution is flawed (that is, future relaxation of documentation requirements for professional billing) because the premise is delusive. Documentation isn’t fundamentally the problem. Having clinicians jump through regulatory hoops which do not advance patients’ care, and providers misunderstanding the requirements for level-of-service billing are the essential issues. Getting no training on how to properly document in medical school/residency and receiving no formative feedback on documentation throughout one’s career compounds the problem. Having clinical documentation serve too many masters, including compliance, quality, medicolegal, utilization review, and reimbursement, is also to blame. The advent of the electronic medical record was just the straw that broke the camel’s back.

Many hospitals now have a clinical documentation integrity (CDI) team which is tasked with querying the provider when the health record documentation is conflicting, imprecise, incomplete, illegible, ambiguous, or inconsistent. They are charged with getting practitioners to associate clinical indicators with diagnoses and to consider removal of diagnoses which do not seem clinically valid from the existing documentation. From this explanation, you might well conclude that the CDI specialist could generate a query on every patient if they were so inclined, and you would be correct. But the goal isn’t to torture the physician – it is to ensure that the medical record is accurately depicting the encounter.

You are not being asked for more documentation by the CDI team; they are entreating you for higher-quality documentation. Let me give you some pointers to ward off queries.

  • Tell the story. The most important goal of documentation is to clinically communicate to other caregivers. Think to yourself: “What would a fellow clinician need to know about this patient to understand why I drew those conclusions or to pick up where I left off?” At 2 a.m., that information, or lack thereof, could literally be a matter of life or death.
  • Tell the truth. Embellishing the record or including invalid diagnoses with the intent to increase the severity of illness resulting in a more favorable diagnosis-related group – the inpatient risk-adjustment system – is considered fraud.
  • You may like the convenience of copy forward, but do you relish reading other people’s copy and paste? Consider doing a documentation time-out. Before you copy and paste yesterday’s assessment and plan, stop and think: “Why is the patient still here? Why are we doing what we are doing?” If you choose to copy and paste, be certain to do mindful editing so the documentation represents the current situation and avoids redundancy. Appropriately editing copy and pasted documentation may prove more time consuming than generating a note de novo.
  • Translate findings into diagnoses using your best medical judgment. One man’s hypotension may be another health care provider’s shock. Coders are not clinical and are not permitted to make inferences. A potassium of 6.7 may be hyperkalemia or it may be spurious – only a clinician may make that determination using their clinical expertise and experience. The coder is not allowed to read your mind. You must explicitly draw the conclusion that a febrile patient with bacteremia, encephalopathy, hypoxemia, and a blood pressure of 85/60 is in septic shock.
  • Uncertain diagnoses (heralded by words such as: likely, possible, probable, suspected, rule out, etc.) which are not ruled out prior to discharge or demise are coded as if they were definitively present, for the inpatient technical side of hospital billing. This is distinctly different than the professional fee where you can only code definitive diagnoses. If you have a strong suspicion (not wild speculation) that a condition is present, best practice is to offer an uncertain diagnosis. Associate signs and symptoms with your most likely diagnosis: “Shortness of breath, pleuritic chest pain, and hypoxemia in the setting of cancer, probable pulmonary embolism.”
  • Evolve, resolve, remove, and recap. If an uncertain diagnosis is ruled in, take away the uncertainty. If it is ruled out, don’t have 4 days of copy and pasted: “Possible eosinophilic pneumonia.” You do not have to maintain a resolved diagnosis ad infinitum. It can drop off the diagnosis list but be sure to have it reappear in the discharge summary.
  • I know it can be a hASSLe to do excellent documentation, but it is critical for many reasons, most importantly for superlative patient care. More accurate coding and billing is an intended consequence. A: Acuity; S: Severity; S: Specificity (may affect the coding and the risk-adjustment implications. Acute systolic heart failure does not equal heart failure; type 2 diabetes mellitus with diabetic chronic kidney disease, stage 4 does not equal chronic kidney disease); and L: Linkage (of diagnosis with underlying cause or manifestation [e.g., because of, associated with, as a result of, secondary to, or from diabetic nephropathy, hypertensive encephalopathy]).
  • If you have the capability to keep a running summary throughout the hospital stay, do so and keep it updated. A few moments of daily careful editing and composing can save time and effort at the back end creating the discharge summary. The follow-up care provider can reconstruct the hospital course and it is your last chance to spin the narrative for the lawyers.
  • Read your documentation over. Ensure that it is clear, accurate, concise, and tells the story and the plans for the patient. Make sure that someone reading the note will know what you were thinking.
  • Set up a program to self-audit documentation where monthly or quarterly, you and your partners mutually review a certain number of records and give each other feedback. Design an assessment tool which rates the quality of documentation elements which your hospital/network/service line values (clarity, copy and paste, complete and specific diagnoses, etc.). You know who the best documenters are. Why do you think their documentation is superior? How can you emulate them?

Finally, answer CDI queries. The CDI specialist is your ally, not your enemy. They want you to get credit for taking care of sick and complex patients. They are not permitted to lead the provider, so don’t ask them what they want you to write. But, if you don’t understand the query or issue, have a conversation and get it clarified. It is in everyone’s best interest to get this right.

Documentation improves patient care and demonstrates that you provided excellent patient care. Put mentation back into documentation.

Dr. Remer was a practicing emergency physician for 25 years and a physician advisor for 4 years. She is on the board of directors of the American College of Physician Advisors and the advisory board of the Association of Clinical Documentation Improvement Specialists. She currently provides consulting services for provider education on documentation, CDI, and ICD-10 coding. Dr. Remer can be reached at eremer@icd10md.com

Documentation has always been part of a physician’s job. Historically, in the days of paper records, physicians saw a patient on rounds and immediately following, while still on the unit, wrote a daily note detailing the events, test results, and plans since the last note. Addenda were written over the course of the day and night as needed.

Dr. Erica Remer, on the board of directors of the American College of Physician Advisors and the advisory board of the Association of Clinical Documentation Improvement Specialists
Dr. Erica Remer

The medical record was a chronological itemization of the encounter. The chart told the patient’s story, hopefully legibly and without excessive rehashing of previous material. The discharge summary then encapsulated the hospitalization in several coherent paragraphs.

In the current electronic records environment, we are inundated with excessive and repetitious information, data without interpretation, differentials without diagnoses. Prepopulation of templated notes, defaults without edit, and dictation without revision have degraded our documentation to the point of unintelligibility. The chronological storytelling and trustworthiness of the medical record has become suspect.

The Centers for Medicare & Medicaid Services is touting its “Patients over Paperwork” initiative. The solution is flawed (that is, future relaxation of documentation requirements for professional billing) because the premise is delusive. Documentation isn’t fundamentally the problem. Having clinicians jump through regulatory hoops which do not advance patients’ care, and providers misunderstanding the requirements for level-of-service billing are the essential issues. Getting no training on how to properly document in medical school/residency and receiving no formative feedback on documentation throughout one’s career compounds the problem. Having clinical documentation serve too many masters, including compliance, quality, medicolegal, utilization review, and reimbursement, is also to blame. The advent of the electronic medical record was just the straw that broke the camel’s back.

Many hospitals now have a clinical documentation integrity (CDI) team which is tasked with querying the provider when the health record documentation is conflicting, imprecise, incomplete, illegible, ambiguous, or inconsistent. They are charged with getting practitioners to associate clinical indicators with diagnoses and to consider removal of diagnoses which do not seem clinically valid from the existing documentation. From this explanation, you might well conclude that the CDI specialist could generate a query on every patient if they were so inclined, and you would be correct. But the goal isn’t to torture the physician – it is to ensure that the medical record is accurately depicting the encounter.

You are not being asked for more documentation by the CDI team; they are entreating you for higher-quality documentation. Let me give you some pointers to ward off queries.

  • Tell the story. The most important goal of documentation is to clinically communicate to other caregivers. Think to yourself: “What would a fellow clinician need to know about this patient to understand why I drew those conclusions or to pick up where I left off?” At 2 a.m., that information, or lack thereof, could literally be a matter of life or death.
  • Tell the truth. Embellishing the record or including invalid diagnoses with the intent to increase the severity of illness resulting in a more favorable diagnosis-related group – the inpatient risk-adjustment system – is considered fraud.
  • You may like the convenience of copy forward, but do you relish reading other people’s copy and paste? Consider doing a documentation time-out. Before you copy and paste yesterday’s assessment and plan, stop and think: “Why is the patient still here? Why are we doing what we are doing?” If you choose to copy and paste, be certain to do mindful editing so the documentation represents the current situation and avoids redundancy. Appropriately editing copy and pasted documentation may prove more time consuming than generating a note de novo.
  • Translate findings into diagnoses using your best medical judgment. One man’s hypotension may be another health care provider’s shock. Coders are not clinical and are not permitted to make inferences. A potassium of 6.7 may be hyperkalemia or it may be spurious – only a clinician may make that determination using their clinical expertise and experience. The coder is not allowed to read your mind. You must explicitly draw the conclusion that a febrile patient with bacteremia, encephalopathy, hypoxemia, and a blood pressure of 85/60 is in septic shock.
  • Uncertain diagnoses (heralded by words such as: likely, possible, probable, suspected, rule out, etc.) which are not ruled out prior to discharge or demise are coded as if they were definitively present, for the inpatient technical side of hospital billing. This is distinctly different than the professional fee where you can only code definitive diagnoses. If you have a strong suspicion (not wild speculation) that a condition is present, best practice is to offer an uncertain diagnosis. Associate signs and symptoms with your most likely diagnosis: “Shortness of breath, pleuritic chest pain, and hypoxemia in the setting of cancer, probable pulmonary embolism.”
  • Evolve, resolve, remove, and recap. If an uncertain diagnosis is ruled in, take away the uncertainty. If it is ruled out, don’t have 4 days of copy and pasted: “Possible eosinophilic pneumonia.” You do not have to maintain a resolved diagnosis ad infinitum. It can drop off the diagnosis list but be sure to have it reappear in the discharge summary.
  • I know it can be a hASSLe to do excellent documentation, but it is critical for many reasons, most importantly for superlative patient care. More accurate coding and billing is an intended consequence. A: Acuity; S: Severity; S: Specificity (may affect the coding and the risk-adjustment implications. Acute systolic heart failure does not equal heart failure; type 2 diabetes mellitus with diabetic chronic kidney disease, stage 4 does not equal chronic kidney disease); and L: Linkage (of diagnosis with underlying cause or manifestation [e.g., because of, associated with, as a result of, secondary to, or from diabetic nephropathy, hypertensive encephalopathy]).
  • If you have the capability to keep a running summary throughout the hospital stay, do so and keep it updated. A few moments of daily careful editing and composing can save time and effort at the back end creating the discharge summary. The follow-up care provider can reconstruct the hospital course and it is your last chance to spin the narrative for the lawyers.
  • Read your documentation over. Ensure that it is clear, accurate, concise, and tells the story and the plans for the patient. Make sure that someone reading the note will know what you were thinking.
  • Set up a program to self-audit documentation where monthly or quarterly, you and your partners mutually review a certain number of records and give each other feedback. Design an assessment tool which rates the quality of documentation elements which your hospital/network/service line values (clarity, copy and paste, complete and specific diagnoses, etc.). You know who the best documenters are. Why do you think their documentation is superior? How can you emulate them?

Finally, answer CDI queries. The CDI specialist is your ally, not your enemy. They want you to get credit for taking care of sick and complex patients. They are not permitted to lead the provider, so don’t ask them what they want you to write. But, if you don’t understand the query or issue, have a conversation and get it clarified. It is in everyone’s best interest to get this right.

Documentation improves patient care and demonstrates that you provided excellent patient care. Put mentation back into documentation.

Dr. Remer was a practicing emergency physician for 25 years and a physician advisor for 4 years. She is on the board of directors of the American College of Physician Advisors and the advisory board of the Association of Clinical Documentation Improvement Specialists. She currently provides consulting services for provider education on documentation, CDI, and ICD-10 coding. Dr. Remer can be reached at eremer@icd10md.com

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.

Value of very early etanercept plus methotrexate not confirmed in real-world RA trial

Article Type
Changed
Tue, 02/04/2020 - 14:19

An open-label, randomized study did not confirm that patients with very early rheumatoid arthritis obtain an outsized benefit from first-line combination treatment with etanercept (Enbrel) plus methotrexate, investigators say.

Dr. Paul Emery, professor of rheumatology and director of the University of Leeds (UK) Musculoskeletal Biomedical Research Center
Bruce Jancin/MDedge News
Dr. Paul Emery

A remission rate of 52% was seen with first-line etanercept plus methotrexate, compared with 38% for a strategy of methotrexate escalated to add etanercept in patients not in remission at 24 weeks in the study, known as VEDERA (Very Early Versus Delayed Etanercept in Patients With RA).

Investigators said a difference of 14 percentage points between remission rates was comparable to what was seen among patients with early RA in an earlier randomized trial of etanercept plus methotrexate versus methotrexate monotherapy.

However, the difference was not on par with the “larger than standard” effect of about 30% seen in an exploratory analysis of the very early RA subset in that previous study, according to VEDERA study authors, led by Paul Emery, MD, of the University of Leeds (England).

Taken together, the results highlight a “ceiling effect” in achieving remission in this real-life, treatment-naive cohort, Dr. Emery and coauthors noted in their report, which appears in Annals of the Rheumatic Diseases.

The study population aligned with real-world clinical practice, according to the investigators, who noted that half the cohort had at least one comorbidity.

“This may have partly driven the generally poorer than expected performance, the exact mechanisms for which are unclear,” they wrote in their discussion of results.

Delaying etanercept until failure of methotrexate, instead of giving both drugs up front, was linked to poorer etanercept response in an exploratory analysis of VEDERA. However, Dr. Emery and coinvestigators noted that this finding “requires validation and further investigation.”

While first-line etanercept plus methotrexate is a “clinically appropriate approach” in early RA, results of the VEDERA study don’t help to inform clinicians as to when it would be prudent to select that therapeutic approach, said Daniel E. Furst, MD, professor of medicine (emeritus) at the University of California, Los Angeles.

Dr. Daniel E. Furst, professor of rheumatology at the University of Washington, Seattle, who also is affiliated with the University of California, Los Angeles, and the University of Florence, Italy.
Dr. Daniel E. Furst

“Whether you’re treating with methotrexate or methotrexate plus etanercept, they do pretty well,” Dr. Furst said in an interview. “What that says to me is when you have patients with very early RA and the disease is moderately active, you should really try methotrexate before you add expensive other drugs.”

The phase 4, open-label, randomized VEDERA trial included 120 adult patients with new-onset early RA with symptom duration of less than 12 months. All patients in VEDERA had a 28-joint disease activity score based on erythrocyte sedimentation rate (DAS28-ESR) of 3.2 or greater and clinical evidence of synovitis. They were all positive for anti-citrullinated peptide antibody or rheumatoid factor, or had evidence of disease activity in at least one joint by power Doppler ultrasonography.

The patients were randomized to 48 weeks of treatment with either first-line etanercept plus methotrexate, or with methotrexate in a treat-to-target strategy that called for the addition of etanercept if the DAS28-ESR was still 2.6 or greater at 24 weeks.

Based on results of the earlier trial, known as COMET, a confirmatory remission rate in the etanercept plus methotrexate arm was anticipated to be 70%, versus 40% for the methotrexate treat-to-target arm, investigators said in a discussion of their statistical methods.

Study results did not confirm a large effect size, according to the investigators. By week 48, DAS28-ESR remission was achieved in 52% of patients in the first-line etanercept plus methotrexate arm, versus 38% in the methotrexate treat-to-target arm, for an absolute difference of 14 percentage points (odds ratio, 1.73; 95% confidence interval, 0.81-3.70; P = .160).



In early, new-onset RA, remission is the goal, Dr. Emery and coauthors said. The proportions of patients in remission in both arms are “suboptimal rates for the contemporary era,” they wrote.

The escalation to etanercept at week 24 did not improve remission rates appreciably, with about 60% still failing to achieve that endpoint. However, an exploratory analysis suggested the subsequent 24 weeks of etanercept exposure in those escalated patients was associated with a lower rate of remission, compared with 24 weeks of etanercept in the front-line approach, investigators said.

In that analysis, the adjusted odds ratio of achieving DAS28-ESR remission was 2.84 (95% CI, 0.84-9.60) in favor of the first-line etanercept approach.

Dr. Furst said in the interview that the VEDERA results are subject to the inherent biases of an open-label study. He also suggested that further investigations could compare the two first-line treatment approaches specifically in very early RA patients with markers of more severe disease.

“That’s the only way I would think we might gain a little bit more, but so far, these data don’t support getting terribly aggressive,” he said.

The study was funded through an investigator-sponsored research grant provided by Pfizer. Three authors disclosed financial relationships with multiple pharmaceutical companies that market drugs for RA, including Pfizer.

SOURCE: Emery P et al. Ann Rheum Dis. 2020 Jan 29. doi: 10.1136/annrheumdis-2019-216539.

Publications
Topics
Sections

An open-label, randomized study did not confirm that patients with very early rheumatoid arthritis obtain an outsized benefit from first-line combination treatment with etanercept (Enbrel) plus methotrexate, investigators say.

Dr. Paul Emery, professor of rheumatology and director of the University of Leeds (UK) Musculoskeletal Biomedical Research Center
Bruce Jancin/MDedge News
Dr. Paul Emery

A remission rate of 52% was seen with first-line etanercept plus methotrexate, compared with 38% for a strategy of methotrexate escalated to add etanercept in patients not in remission at 24 weeks in the study, known as VEDERA (Very Early Versus Delayed Etanercept in Patients With RA).

Investigators said a difference of 14 percentage points between remission rates was comparable to what was seen among patients with early RA in an earlier randomized trial of etanercept plus methotrexate versus methotrexate monotherapy.

However, the difference was not on par with the “larger than standard” effect of about 30% seen in an exploratory analysis of the very early RA subset in that previous study, according to VEDERA study authors, led by Paul Emery, MD, of the University of Leeds (England).

Taken together, the results highlight a “ceiling effect” in achieving remission in this real-life, treatment-naive cohort, Dr. Emery and coauthors noted in their report, which appears in Annals of the Rheumatic Diseases.

The study population aligned with real-world clinical practice, according to the investigators, who noted that half the cohort had at least one comorbidity.

“This may have partly driven the generally poorer than expected performance, the exact mechanisms for which are unclear,” they wrote in their discussion of results.

Delaying etanercept until failure of methotrexate, instead of giving both drugs up front, was linked to poorer etanercept response in an exploratory analysis of VEDERA. However, Dr. Emery and coinvestigators noted that this finding “requires validation and further investigation.”

While first-line etanercept plus methotrexate is a “clinically appropriate approach” in early RA, results of the VEDERA study don’t help to inform clinicians as to when it would be prudent to select that therapeutic approach, said Daniel E. Furst, MD, professor of medicine (emeritus) at the University of California, Los Angeles.

Dr. Daniel E. Furst, professor of rheumatology at the University of Washington, Seattle, who also is affiliated with the University of California, Los Angeles, and the University of Florence, Italy.
Dr. Daniel E. Furst

“Whether you’re treating with methotrexate or methotrexate plus etanercept, they do pretty well,” Dr. Furst said in an interview. “What that says to me is when you have patients with very early RA and the disease is moderately active, you should really try methotrexate before you add expensive other drugs.”

The phase 4, open-label, randomized VEDERA trial included 120 adult patients with new-onset early RA with symptom duration of less than 12 months. All patients in VEDERA had a 28-joint disease activity score based on erythrocyte sedimentation rate (DAS28-ESR) of 3.2 or greater and clinical evidence of synovitis. They were all positive for anti-citrullinated peptide antibody or rheumatoid factor, or had evidence of disease activity in at least one joint by power Doppler ultrasonography.

The patients were randomized to 48 weeks of treatment with either first-line etanercept plus methotrexate, or with methotrexate in a treat-to-target strategy that called for the addition of etanercept if the DAS28-ESR was still 2.6 or greater at 24 weeks.

Based on results of the earlier trial, known as COMET, a confirmatory remission rate in the etanercept plus methotrexate arm was anticipated to be 70%, versus 40% for the methotrexate treat-to-target arm, investigators said in a discussion of their statistical methods.

Study results did not confirm a large effect size, according to the investigators. By week 48, DAS28-ESR remission was achieved in 52% of patients in the first-line etanercept plus methotrexate arm, versus 38% in the methotrexate treat-to-target arm, for an absolute difference of 14 percentage points (odds ratio, 1.73; 95% confidence interval, 0.81-3.70; P = .160).



In early, new-onset RA, remission is the goal, Dr. Emery and coauthors said. The proportions of patients in remission in both arms are “suboptimal rates for the contemporary era,” they wrote.

The escalation to etanercept at week 24 did not improve remission rates appreciably, with about 60% still failing to achieve that endpoint. However, an exploratory analysis suggested the subsequent 24 weeks of etanercept exposure in those escalated patients was associated with a lower rate of remission, compared with 24 weeks of etanercept in the front-line approach, investigators said.

In that analysis, the adjusted odds ratio of achieving DAS28-ESR remission was 2.84 (95% CI, 0.84-9.60) in favor of the first-line etanercept approach.

Dr. Furst said in the interview that the VEDERA results are subject to the inherent biases of an open-label study. He also suggested that further investigations could compare the two first-line treatment approaches specifically in very early RA patients with markers of more severe disease.

“That’s the only way I would think we might gain a little bit more, but so far, these data don’t support getting terribly aggressive,” he said.

The study was funded through an investigator-sponsored research grant provided by Pfizer. Three authors disclosed financial relationships with multiple pharmaceutical companies that market drugs for RA, including Pfizer.

SOURCE: Emery P et al. Ann Rheum Dis. 2020 Jan 29. doi: 10.1136/annrheumdis-2019-216539.

An open-label, randomized study did not confirm that patients with very early rheumatoid arthritis obtain an outsized benefit from first-line combination treatment with etanercept (Enbrel) plus methotrexate, investigators say.

Dr. Paul Emery, professor of rheumatology and director of the University of Leeds (UK) Musculoskeletal Biomedical Research Center
Bruce Jancin/MDedge News
Dr. Paul Emery

A remission rate of 52% was seen with first-line etanercept plus methotrexate, compared with 38% for a strategy of methotrexate escalated to add etanercept in patients not in remission at 24 weeks in the study, known as VEDERA (Very Early Versus Delayed Etanercept in Patients With RA).

Investigators said a difference of 14 percentage points between remission rates was comparable to what was seen among patients with early RA in an earlier randomized trial of etanercept plus methotrexate versus methotrexate monotherapy.

However, the difference was not on par with the “larger than standard” effect of about 30% seen in an exploratory analysis of the very early RA subset in that previous study, according to VEDERA study authors, led by Paul Emery, MD, of the University of Leeds (England).

Taken together, the results highlight a “ceiling effect” in achieving remission in this real-life, treatment-naive cohort, Dr. Emery and coauthors noted in their report, which appears in Annals of the Rheumatic Diseases.

The study population aligned with real-world clinical practice, according to the investigators, who noted that half the cohort had at least one comorbidity.

“This may have partly driven the generally poorer than expected performance, the exact mechanisms for which are unclear,” they wrote in their discussion of results.

Delaying etanercept until failure of methotrexate, instead of giving both drugs up front, was linked to poorer etanercept response in an exploratory analysis of VEDERA. However, Dr. Emery and coinvestigators noted that this finding “requires validation and further investigation.”

While first-line etanercept plus methotrexate is a “clinically appropriate approach” in early RA, results of the VEDERA study don’t help to inform clinicians as to when it would be prudent to select that therapeutic approach, said Daniel E. Furst, MD, professor of medicine (emeritus) at the University of California, Los Angeles.

Dr. Daniel E. Furst, professor of rheumatology at the University of Washington, Seattle, who also is affiliated with the University of California, Los Angeles, and the University of Florence, Italy.
Dr. Daniel E. Furst

“Whether you’re treating with methotrexate or methotrexate plus etanercept, they do pretty well,” Dr. Furst said in an interview. “What that says to me is when you have patients with very early RA and the disease is moderately active, you should really try methotrexate before you add expensive other drugs.”

The phase 4, open-label, randomized VEDERA trial included 120 adult patients with new-onset early RA with symptom duration of less than 12 months. All patients in VEDERA had a 28-joint disease activity score based on erythrocyte sedimentation rate (DAS28-ESR) of 3.2 or greater and clinical evidence of synovitis. They were all positive for anti-citrullinated peptide antibody or rheumatoid factor, or had evidence of disease activity in at least one joint by power Doppler ultrasonography.

The patients were randomized to 48 weeks of treatment with either first-line etanercept plus methotrexate, or with methotrexate in a treat-to-target strategy that called for the addition of etanercept if the DAS28-ESR was still 2.6 or greater at 24 weeks.

Based on results of the earlier trial, known as COMET, a confirmatory remission rate in the etanercept plus methotrexate arm was anticipated to be 70%, versus 40% for the methotrexate treat-to-target arm, investigators said in a discussion of their statistical methods.

Study results did not confirm a large effect size, according to the investigators. By week 48, DAS28-ESR remission was achieved in 52% of patients in the first-line etanercept plus methotrexate arm, versus 38% in the methotrexate treat-to-target arm, for an absolute difference of 14 percentage points (odds ratio, 1.73; 95% confidence interval, 0.81-3.70; P = .160).



In early, new-onset RA, remission is the goal, Dr. Emery and coauthors said. The proportions of patients in remission in both arms are “suboptimal rates for the contemporary era,” they wrote.

The escalation to etanercept at week 24 did not improve remission rates appreciably, with about 60% still failing to achieve that endpoint. However, an exploratory analysis suggested the subsequent 24 weeks of etanercept exposure in those escalated patients was associated with a lower rate of remission, compared with 24 weeks of etanercept in the front-line approach, investigators said.

In that analysis, the adjusted odds ratio of achieving DAS28-ESR remission was 2.84 (95% CI, 0.84-9.60) in favor of the first-line etanercept approach.

Dr. Furst said in the interview that the VEDERA results are subject to the inherent biases of an open-label study. He also suggested that further investigations could compare the two first-line treatment approaches specifically in very early RA patients with markers of more severe disease.

“That’s the only way I would think we might gain a little bit more, but so far, these data don’t support getting terribly aggressive,” he said.

The study was funded through an investigator-sponsored research grant provided by Pfizer. Three authors disclosed financial relationships with multiple pharmaceutical companies that market drugs for RA, including Pfizer.

SOURCE: Emery P et al. Ann Rheum Dis. 2020 Jan 29. doi: 10.1136/annrheumdis-2019-216539.

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

FROM ANNALS OF THE RHEUMATIC DISEASES

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.

Hypertensive disorders of pregnancy in SLE contribute to later CV outcomes

Article Type
Changed
Fri, 02/07/2020 - 11:15

Women with systemic lupus erythematosus (SLE) who experience hypertensive disorders of pregnancy may have a higher rate of cardiovascular outcomes after pregnancy, as well as a higher rate of hypertension later in life, than do those without maternal hypertension, according to findings from a Swedish population-based, longitudinal cohort study.

“Premature CVD [cardiovascular disease] is a well-documented complication in women with SLE, which is likely, at least in part, due to renal disease, prothrombotic [antiphospholipid antibodies], and systemic inflammation. Our data confirm that women who experience a hypertensive disorder in pregnancy [HDP] are at greater risk of developing hypertension after pregnancy, and that this association is also evident for women with SLE. Women with SLE and HDP were also at increased risk of CVD, particularly stroke, at young ages and should be monitored closely and consider treatment to attenuate risk,” wrote first author Julia F. Simard, ScD, of Stanford (Calif.) University and colleagues in Arthritis Care & Research.

To reach those conclusions, the researchers identified 3,340 women in the Swedish Medical Birth Register with their first singleton delivery during 1987-2012. They matched each of the 450 women with prevalent SLE from the Medical Birth Register to 5 women without SLE in the National Patient Register based on sex, birth year, calendar time, and county of residence.



During a median follow-up period of nearly 11 years, women with SLE had an unadjusted incidence rate of incident cardiovascular outcomes of 50 cases per 10,000 person-years versus 7.2 for women without SLE. Cardiovascular outcomes included fatal and nonfatal acute MI, fatal and nonfatal stroke, transient ischemic attacks, unstable angina, and heart failure. A history of HDP in women with SLE, including preeclampsia, was linked with about a twofold higher rate of cardiovascular outcomes regardless of multiple sensitivity analyses, both before and after adjusting for maternal age at delivery, county of birth, education, body mass index, and first-trimester smoking.

The researchers found that the hazard ratio for cardiovascular outcomes in women with SLE and HDP was about eight times higher than the hazard ratio for women without SLE but with HDP, but the relative rarity of cardiovascular events seen during the follow-up period, particularly among women without SLE, made it so that they “could not confirm established associations between HDP and CVD, possibly due to the relatively short follow-up time given that premenopausal CVD is rare among women free of SLE.”

HDP was associated with a threefold higher risk for incident hypertension later in life regardless of SLE status, even though the unadjusted incidence rate was 524 cases per 10,000 person-years among women with both SLE and HDP, compared with 177 per 10,000 person-years among women with HDP in the general population, which sensitivity analyses suggested “was not due to misclassification of antihypertensive use for renal disease in women with SLE nor antihypertensive use for possible HDP in subsequent pregnancies,” the researchers wrote.

Several authors reported research grants from the National Institutes of Health, the Karolinska Institute, the Swedish Research Council, Swedish Heart-Lung Foundation, Stockholm County Council, the King Gustaf V 80th Birthday Fund, the Swedish Rheumatism Association, and Ingegerd Johansson’s Foundation that helped to fund the study. All authors reported having no competing interests.

SOURCE: Simard JF et al. Arthritis Care Res. 2020 Jan 31. doi: 10.1002/acr.24160.

Publications
Topics
Sections

Women with systemic lupus erythematosus (SLE) who experience hypertensive disorders of pregnancy may have a higher rate of cardiovascular outcomes after pregnancy, as well as a higher rate of hypertension later in life, than do those without maternal hypertension, according to findings from a Swedish population-based, longitudinal cohort study.

“Premature CVD [cardiovascular disease] is a well-documented complication in women with SLE, which is likely, at least in part, due to renal disease, prothrombotic [antiphospholipid antibodies], and systemic inflammation. Our data confirm that women who experience a hypertensive disorder in pregnancy [HDP] are at greater risk of developing hypertension after pregnancy, and that this association is also evident for women with SLE. Women with SLE and HDP were also at increased risk of CVD, particularly stroke, at young ages and should be monitored closely and consider treatment to attenuate risk,” wrote first author Julia F. Simard, ScD, of Stanford (Calif.) University and colleagues in Arthritis Care & Research.

To reach those conclusions, the researchers identified 3,340 women in the Swedish Medical Birth Register with their first singleton delivery during 1987-2012. They matched each of the 450 women with prevalent SLE from the Medical Birth Register to 5 women without SLE in the National Patient Register based on sex, birth year, calendar time, and county of residence.



During a median follow-up period of nearly 11 years, women with SLE had an unadjusted incidence rate of incident cardiovascular outcomes of 50 cases per 10,000 person-years versus 7.2 for women without SLE. Cardiovascular outcomes included fatal and nonfatal acute MI, fatal and nonfatal stroke, transient ischemic attacks, unstable angina, and heart failure. A history of HDP in women with SLE, including preeclampsia, was linked with about a twofold higher rate of cardiovascular outcomes regardless of multiple sensitivity analyses, both before and after adjusting for maternal age at delivery, county of birth, education, body mass index, and first-trimester smoking.

The researchers found that the hazard ratio for cardiovascular outcomes in women with SLE and HDP was about eight times higher than the hazard ratio for women without SLE but with HDP, but the relative rarity of cardiovascular events seen during the follow-up period, particularly among women without SLE, made it so that they “could not confirm established associations between HDP and CVD, possibly due to the relatively short follow-up time given that premenopausal CVD is rare among women free of SLE.”

HDP was associated with a threefold higher risk for incident hypertension later in life regardless of SLE status, even though the unadjusted incidence rate was 524 cases per 10,000 person-years among women with both SLE and HDP, compared with 177 per 10,000 person-years among women with HDP in the general population, which sensitivity analyses suggested “was not due to misclassification of antihypertensive use for renal disease in women with SLE nor antihypertensive use for possible HDP in subsequent pregnancies,” the researchers wrote.

Several authors reported research grants from the National Institutes of Health, the Karolinska Institute, the Swedish Research Council, Swedish Heart-Lung Foundation, Stockholm County Council, the King Gustaf V 80th Birthday Fund, the Swedish Rheumatism Association, and Ingegerd Johansson’s Foundation that helped to fund the study. All authors reported having no competing interests.

SOURCE: Simard JF et al. Arthritis Care Res. 2020 Jan 31. doi: 10.1002/acr.24160.

Women with systemic lupus erythematosus (SLE) who experience hypertensive disorders of pregnancy may have a higher rate of cardiovascular outcomes after pregnancy, as well as a higher rate of hypertension later in life, than do those without maternal hypertension, according to findings from a Swedish population-based, longitudinal cohort study.

“Premature CVD [cardiovascular disease] is a well-documented complication in women with SLE, which is likely, at least in part, due to renal disease, prothrombotic [antiphospholipid antibodies], and systemic inflammation. Our data confirm that women who experience a hypertensive disorder in pregnancy [HDP] are at greater risk of developing hypertension after pregnancy, and that this association is also evident for women with SLE. Women with SLE and HDP were also at increased risk of CVD, particularly stroke, at young ages and should be monitored closely and consider treatment to attenuate risk,” wrote first author Julia F. Simard, ScD, of Stanford (Calif.) University and colleagues in Arthritis Care & Research.

To reach those conclusions, the researchers identified 3,340 women in the Swedish Medical Birth Register with their first singleton delivery during 1987-2012. They matched each of the 450 women with prevalent SLE from the Medical Birth Register to 5 women without SLE in the National Patient Register based on sex, birth year, calendar time, and county of residence.



During a median follow-up period of nearly 11 years, women with SLE had an unadjusted incidence rate of incident cardiovascular outcomes of 50 cases per 10,000 person-years versus 7.2 for women without SLE. Cardiovascular outcomes included fatal and nonfatal acute MI, fatal and nonfatal stroke, transient ischemic attacks, unstable angina, and heart failure. A history of HDP in women with SLE, including preeclampsia, was linked with about a twofold higher rate of cardiovascular outcomes regardless of multiple sensitivity analyses, both before and after adjusting for maternal age at delivery, county of birth, education, body mass index, and first-trimester smoking.

The researchers found that the hazard ratio for cardiovascular outcomes in women with SLE and HDP was about eight times higher than the hazard ratio for women without SLE but with HDP, but the relative rarity of cardiovascular events seen during the follow-up period, particularly among women without SLE, made it so that they “could not confirm established associations between HDP and CVD, possibly due to the relatively short follow-up time given that premenopausal CVD is rare among women free of SLE.”

HDP was associated with a threefold higher risk for incident hypertension later in life regardless of SLE status, even though the unadjusted incidence rate was 524 cases per 10,000 person-years among women with both SLE and HDP, compared with 177 per 10,000 person-years among women with HDP in the general population, which sensitivity analyses suggested “was not due to misclassification of antihypertensive use for renal disease in women with SLE nor antihypertensive use for possible HDP in subsequent pregnancies,” the researchers wrote.

Several authors reported research grants from the National Institutes of Health, the Karolinska Institute, the Swedish Research Council, Swedish Heart-Lung Foundation, Stockholm County Council, the King Gustaf V 80th Birthday Fund, the Swedish Rheumatism Association, and Ingegerd Johansson’s Foundation that helped to fund the study. All authors reported having no competing interests.

SOURCE: Simard JF et al. Arthritis Care Res. 2020 Jan 31. doi: 10.1002/acr.24160.

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

FROM ARTHRITIS CARE & RESEARCH

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.

FDA approves novel pandemic influenza vaccine

Article Type
Changed
Mon, 03/22/2021 - 14:08

The Food and Drug Administration has approved the first and only adjuvanted, cell-based pandemic vaccine to provide active immunization against the influenza A virus H5N1 strain.
 

Influenza A (H5N1) monovalent vaccine, adjuvanted (Audenz, Seqirus) is for use in individuals aged 6 months and older.  It’s designed to be rapidly deployed to help protect the U.S. population and can be stockpiled for first responders in the event of a pandemic.

The vaccine and formulated prefilled syringes used in the vaccine are produced in a state-of-the-art production facility built and supported through a multiyear public-private partnership between Seqirus and the Biomedical Advanced Research and Development Authority (BARDA), part of the Office of the Assistant Secretary for Preparedness and Response at the U.S. Department of Health & Human Services.

“Pandemic influenza viruses can be deadly and spread rapidly, making production of safe, effective vaccines essential in saving lives,” BARDA Director Rick Bright, PhD, said in a company news release.

“With this licensure – the latest FDA-approved vaccine to prevent H5N1 influenza — we celebrate a decade-long partnership to achieve health security goals set by the National Strategy for Pandemic Influenza and the 2019 Executive Order to speed the availability of influenza vaccine. Ultimately, this latest licensure means we can protect more people in an influenza pandemic,” said Bright.

“The approval of Audenz represents a key advance in influenza prevention and pandemic preparedness, combining leading-edge, cell-based manufacturing and adjuvant technologies,” Russell Basser, MD, chief scientist and senior vice president of research and development at Seqirus, said in the news release. “This pandemic influenza vaccine exemplifies our commitment to developing innovative technologies that can help provide rapid response during a pandemic emergency.”

Audenz had FDA fast track designation, a process designed to facilitate the development and expedite the review of drugs to treat serious conditions and fill an unmet medical need.
 

This article first appeared on Medscape.com.

Publications
Topics
Sections

The Food and Drug Administration has approved the first and only adjuvanted, cell-based pandemic vaccine to provide active immunization against the influenza A virus H5N1 strain.
 

Influenza A (H5N1) monovalent vaccine, adjuvanted (Audenz, Seqirus) is for use in individuals aged 6 months and older.  It’s designed to be rapidly deployed to help protect the U.S. population and can be stockpiled for first responders in the event of a pandemic.

The vaccine and formulated prefilled syringes used in the vaccine are produced in a state-of-the-art production facility built and supported through a multiyear public-private partnership between Seqirus and the Biomedical Advanced Research and Development Authority (BARDA), part of the Office of the Assistant Secretary for Preparedness and Response at the U.S. Department of Health & Human Services.

“Pandemic influenza viruses can be deadly and spread rapidly, making production of safe, effective vaccines essential in saving lives,” BARDA Director Rick Bright, PhD, said in a company news release.

“With this licensure – the latest FDA-approved vaccine to prevent H5N1 influenza — we celebrate a decade-long partnership to achieve health security goals set by the National Strategy for Pandemic Influenza and the 2019 Executive Order to speed the availability of influenza vaccine. Ultimately, this latest licensure means we can protect more people in an influenza pandemic,” said Bright.

“The approval of Audenz represents a key advance in influenza prevention and pandemic preparedness, combining leading-edge, cell-based manufacturing and adjuvant technologies,” Russell Basser, MD, chief scientist and senior vice president of research and development at Seqirus, said in the news release. “This pandemic influenza vaccine exemplifies our commitment to developing innovative technologies that can help provide rapid response during a pandemic emergency.”

Audenz had FDA fast track designation, a process designed to facilitate the development and expedite the review of drugs to treat serious conditions and fill an unmet medical need.
 

This article first appeared on Medscape.com.

The Food and Drug Administration has approved the first and only adjuvanted, cell-based pandemic vaccine to provide active immunization against the influenza A virus H5N1 strain.
 

Influenza A (H5N1) monovalent vaccine, adjuvanted (Audenz, Seqirus) is for use in individuals aged 6 months and older.  It’s designed to be rapidly deployed to help protect the U.S. population and can be stockpiled for first responders in the event of a pandemic.

The vaccine and formulated prefilled syringes used in the vaccine are produced in a state-of-the-art production facility built and supported through a multiyear public-private partnership between Seqirus and the Biomedical Advanced Research and Development Authority (BARDA), part of the Office of the Assistant Secretary for Preparedness and Response at the U.S. Department of Health & Human Services.

“Pandemic influenza viruses can be deadly and spread rapidly, making production of safe, effective vaccines essential in saving lives,” BARDA Director Rick Bright, PhD, said in a company news release.

“With this licensure – the latest FDA-approved vaccine to prevent H5N1 influenza — we celebrate a decade-long partnership to achieve health security goals set by the National Strategy for Pandemic Influenza and the 2019 Executive Order to speed the availability of influenza vaccine. Ultimately, this latest licensure means we can protect more people in an influenza pandemic,” said Bright.

“The approval of Audenz represents a key advance in influenza prevention and pandemic preparedness, combining leading-edge, cell-based manufacturing and adjuvant technologies,” Russell Basser, MD, chief scientist and senior vice president of research and development at Seqirus, said in the news release. “This pandemic influenza vaccine exemplifies our commitment to developing innovative technologies that can help provide rapid response during a pandemic emergency.”

Audenz had FDA fast track designation, a process designed to facilitate the development and expedite the review of drugs to treat serious conditions and fill an unmet medical need.
 

This article first appeared on Medscape.com.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Medscape Article

Preoperative CT shows little value in early vulvar SCC

Article Type
Changed
Tue, 02/04/2020 - 12:57

Preoperative computerized tomography (CT) demonstrated limited value in the clinical management of patients with early-stage vulvar squamous cell carcinoma (VSCC) analyzed in a single-center study.

The findings suggest preoperative CT imaging could be excluded prior to sentinel inguinal lymph node biopsy or staging surgery in patients with early-stage disease.

“In this study, we aimed to investigate if preoperative CT scan influences the overall course of VSCC management in patients without clinical evidence of groin lymphadenopathy,” wrote Rachel Pounds, MD, of the University of Birmingham (England) and colleagues. The study was published in Gynecologic Oncology.

The researchers prospectively studied a cohort of 225 patients with primary or recurrent VSCC who underwent staging surgery at a single institution in the United Kingdom. The patients’ mean age was 67 years (range, 54-79 years), and most had stage 1B disease (57.8%).

The researchers compared preoperative imaging findings with histological results from sentinel inguinal lymph node biopsy. Other clinical information, including surgery type, evidence of groin node involvement, and age at diagnosis was collected from patient files and included in the analysis.

In all, 51.6% of patients underwent preoperative CT imaging. Among these patients, 37.9% had a positive report of radiological groin lymphatic metastases.

“True groin node metastases, confirmed histologically, were observed in 26 patients (22.4%) with a radiologically positive scan report (true positives) and in 18 patients (15.5%) with a radiological negative scan report (false negatives),” the researchers wrote.

The specificity and sensitivity of preoperative CT to detect groin lymphatic metastasis were 77.8% and 59.1%, respectively. The positive and negative predictive values were 61.9% and 75.7%, respectively.

There was no significant difference in overall survival, disease-specific or disease-free survival, or groin node recurrence between patients who underwent preoperative CT and patients who did not.

Groin node recurrence was observed in 10.3% of patients with preoperative CT and 11.5% of patients without it (P = .7768). Disease-specific death occurred in 16.4% of patients with preoperative CT and 13.5% of patients without it (P = .5451).

“Our results highlight the poor reliability of preoperative CT scans in detecting inguinal lymphatic metastasis,” the researchers wrote. “Preoperative CT scan may be omitted in early stage VSCC prior to surgical staging as it does not affect overall management and surgical outcomes.”

No funding sources were reported for this study. The authors reported having no conflicts of interest.

SOURCE: Pounds R et al. Gynecol Oncol. 2020 Jan 24. doi: 10.1016/j.ygyno.2020.01.031.

Publications
Topics
Sections

Preoperative computerized tomography (CT) demonstrated limited value in the clinical management of patients with early-stage vulvar squamous cell carcinoma (VSCC) analyzed in a single-center study.

The findings suggest preoperative CT imaging could be excluded prior to sentinel inguinal lymph node biopsy or staging surgery in patients with early-stage disease.

“In this study, we aimed to investigate if preoperative CT scan influences the overall course of VSCC management in patients without clinical evidence of groin lymphadenopathy,” wrote Rachel Pounds, MD, of the University of Birmingham (England) and colleagues. The study was published in Gynecologic Oncology.

The researchers prospectively studied a cohort of 225 patients with primary or recurrent VSCC who underwent staging surgery at a single institution in the United Kingdom. The patients’ mean age was 67 years (range, 54-79 years), and most had stage 1B disease (57.8%).

The researchers compared preoperative imaging findings with histological results from sentinel inguinal lymph node biopsy. Other clinical information, including surgery type, evidence of groin node involvement, and age at diagnosis was collected from patient files and included in the analysis.

In all, 51.6% of patients underwent preoperative CT imaging. Among these patients, 37.9% had a positive report of radiological groin lymphatic metastases.

“True groin node metastases, confirmed histologically, were observed in 26 patients (22.4%) with a radiologically positive scan report (true positives) and in 18 patients (15.5%) with a radiological negative scan report (false negatives),” the researchers wrote.

The specificity and sensitivity of preoperative CT to detect groin lymphatic metastasis were 77.8% and 59.1%, respectively. The positive and negative predictive values were 61.9% and 75.7%, respectively.

There was no significant difference in overall survival, disease-specific or disease-free survival, or groin node recurrence between patients who underwent preoperative CT and patients who did not.

Groin node recurrence was observed in 10.3% of patients with preoperative CT and 11.5% of patients without it (P = .7768). Disease-specific death occurred in 16.4% of patients with preoperative CT and 13.5% of patients without it (P = .5451).

“Our results highlight the poor reliability of preoperative CT scans in detecting inguinal lymphatic metastasis,” the researchers wrote. “Preoperative CT scan may be omitted in early stage VSCC prior to surgical staging as it does not affect overall management and surgical outcomes.”

No funding sources were reported for this study. The authors reported having no conflicts of interest.

SOURCE: Pounds R et al. Gynecol Oncol. 2020 Jan 24. doi: 10.1016/j.ygyno.2020.01.031.

Preoperative computerized tomography (CT) demonstrated limited value in the clinical management of patients with early-stage vulvar squamous cell carcinoma (VSCC) analyzed in a single-center study.

The findings suggest preoperative CT imaging could be excluded prior to sentinel inguinal lymph node biopsy or staging surgery in patients with early-stage disease.

“In this study, we aimed to investigate if preoperative CT scan influences the overall course of VSCC management in patients without clinical evidence of groin lymphadenopathy,” wrote Rachel Pounds, MD, of the University of Birmingham (England) and colleagues. The study was published in Gynecologic Oncology.

The researchers prospectively studied a cohort of 225 patients with primary or recurrent VSCC who underwent staging surgery at a single institution in the United Kingdom. The patients’ mean age was 67 years (range, 54-79 years), and most had stage 1B disease (57.8%).

The researchers compared preoperative imaging findings with histological results from sentinel inguinal lymph node biopsy. Other clinical information, including surgery type, evidence of groin node involvement, and age at diagnosis was collected from patient files and included in the analysis.

In all, 51.6% of patients underwent preoperative CT imaging. Among these patients, 37.9% had a positive report of radiological groin lymphatic metastases.

“True groin node metastases, confirmed histologically, were observed in 26 patients (22.4%) with a radiologically positive scan report (true positives) and in 18 patients (15.5%) with a radiological negative scan report (false negatives),” the researchers wrote.

The specificity and sensitivity of preoperative CT to detect groin lymphatic metastasis were 77.8% and 59.1%, respectively. The positive and negative predictive values were 61.9% and 75.7%, respectively.

There was no significant difference in overall survival, disease-specific or disease-free survival, or groin node recurrence between patients who underwent preoperative CT and patients who did not.

Groin node recurrence was observed in 10.3% of patients with preoperative CT and 11.5% of patients without it (P = .7768). Disease-specific death occurred in 16.4% of patients with preoperative CT and 13.5% of patients without it (P = .5451).

“Our results highlight the poor reliability of preoperative CT scans in detecting inguinal lymphatic metastasis,” the researchers wrote. “Preoperative CT scan may be omitted in early stage VSCC prior to surgical staging as it does not affect overall management and surgical outcomes.”

No funding sources were reported for this study. The authors reported having no conflicts of interest.

SOURCE: Pounds R et al. Gynecol Oncol. 2020 Jan 24. doi: 10.1016/j.ygyno.2020.01.031.

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM GYNECOLOGIC ONCOLOGY

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.

Rate of suicide is higher in people with neurologic disorders

Article Type
Changed
Thu, 12/15/2022 - 15:45

People with neurologic disorders have a higher rate of suicide, compared with people without neurologic conditions, according to a population-based study in Denmark.

The absolute risk difference is small, but statistically significant. “These findings do not necessarily warrant changing the management of treatment for individual patients,” wrote Annette Erlangsen, PhD, a researcher at the Danish Research Institute for Suicide Prevention in Hellerup, and colleagues. “As with all patients, physicians should be aware of the potential for depression, demoralization, and suicide.”

In addition, dementia, Alzheimer’s disease, and intellectual disabilities may be associated with lower suicide rates, according to the study, which was published in JAMA.

“Plausible mechanisms” could underlie the association between neurologic disease and suicide, the authors wrote. A neurologic diagnosis “may constitute a distressing life event,” and the diseases may have psychological, physical, and psychiatric effects. Patients may see themselves as a burden or have less financial security. In addition, the diseases may entail “communication difficulties, poor sleep, and pain.” Neurologic diseases may alter brain circuitry and functioning and influence aggression and impulsivity. “People with neurologic disorders may also have easier access to toxic medication,” they added.
 

More than a dozen conditions examined

Prior studies have found associations between neurologic conditions and rates of suicide, but data have been inconclusive or inconsistent for some of the disorders. To examine whether people with neurologic disorders have higher suicide rates, relative to people without these disorders, the researchers conducted a retrospective study. They analyzed data from more than 7.3 million people aged 15 years or older who lived in Denmark between 1980 and 2016. The cohort included more than 1.2 million people with neurologic disorders. The investigators identified neurologic disorders using ICD codes for head injury, stroke, epilepsy, polyneuropathy, diseases of the myoneural junction, Parkinson’s disease, multiple sclerosis, CNS infections, meningitis, encephalitis, amyotrophic lateral sclerosis, Huntington’s disease, dementia, intellectual disability, and other brain disorders. They compared incidence rates using a Poisson regression model and adjusted for time period, sex, age, region, socioeconomic status, comorbidity, self-harm or psychiatric hospitalization prior to a neurologic diagnosis, and whether a person lived alone.

In all, 35,483 people in the cohort died by suicide at an average age of about 52 years; 77.4% were male. About 15% of those who died by suicide had a neurologic disorder. The suicide incidence rate among people with a neurologic disorder was 44.0 per 100,000 person-years, whereas the rate among people without a neurologic disorder was 20.1 per 100,000 person-years.

The adjusted incidence rate ratio for people with a neurologic disorder was 1.8. The rate ratio was highest during the 3 months after diagnosis, at 3.1. Huntington’s disease and amyotrophic lateral sclerosis were associated with “the largest excess adjusted [incidence rate ratios] of suicide mortality,” with a rate ratio of 4.9 for each condition, the researchers reported. The adjusted incidence rate ratio was 1.7 for head injury, 1.3 for stroke, 1.7 for epilepsy, 1.4 for intracerebral hemorrhage, 1.3 for cerebral infarction, 1.3 for subarachnoid hemorrhage, 1.7 for polyneuropathy and peripheral neuropathy, 2.2 for Guillain-Barré syndrome, 1.9 for diseases of myoneural junction and muscle, 1.8 for other brain disorders, 1.7 for Parkinson’s disease, 2.2 for multiple sclerosis, and 1.6 for CNS infection.

Compared with people without a neurologic condition, people with dementia, Alzheimer’s disease, and intellectual disabilities had lower suicide rates, with adjusted incidence rate ratios of 0.8, 0.2, and 0.6, respectively. “However, the adjusted [incidence rate ratio] for people with dementia during the first month after diagnosis was 3.0,” the researchers wrote.

In addition, the suicide rate increased with an increasing cumulative number of hospital contacts for neurologic conditions.

 

 



Overall incidence rates declined

“Over the study period, the suicide incidence rate for people with neurological disorders decreased from 78.6 per 100,000 person-years during the 1980-1999 years to 27.3 per 100,000 person-years during the 2000-2016 years,” wrote Dr. Erlangsen and colleagues. “The suicide incidence rate for those without a disorder decreased from 26.3 to 12.7 during the same time spans. ... The decline in the overall suicide rate over time did not affect the relative risk pattern.”

The decline in the general suicide rate in Denmark “has largely been attributed to means restriction, such as efforts to limit availability of firearms and particularly toxic medication,” the authors added.

In those time spans, the adjusted incidence rate ratio for suicide among those with dementia decreased from 2.4 to 1.0, and among those with multiple sclerosis from 2.0 to 1.0. “It is possible that the improvements observed for dementia and multiple sclerosis may be related to improvements in treatment and intensified community-based support,” Dr. Erlangsen and coauthors wrote.

When the researchers used people with rheumatoid arthritis as a reference group, those with a neurologic disorder had a higher suicide rate per 100,000 person-years, 30.2 versus 18.4. The adjusted incidence rate ratio for that comparison was 1.4.

In patients with Huntington’s disease, depression mediated by hyperactivity in the hypothalamic-pituitary-adrenal axis may contribute to the risk of suicide. “Witnessing the course of the disease in one’s parent” also may contribute the risk, the researchers wrote.

The analysis may have missed people with neurologic disorders diagnosed before 1977 if they did not have subsequent contact with a hospital, the investigators noted. In addition, diagnoses given in primary care were not included, suicide deaths may be underrecorded, and “adjusting for preexisting mental disorders could be viewed as overadjusting,” they wrote.

The study was supported by a grant from the Psychiatric Research Foundation in Denmark. The authors reported that they had no disclosures.

SOURCE: Erlangsen A et al. JAMA. 2020 Feb 4. doi: 10.1001/jama.2019.21834.

Issue
Neurology Reviews- 28(3)
Publications
Topics
Sections

People with neurologic disorders have a higher rate of suicide, compared with people without neurologic conditions, according to a population-based study in Denmark.

The absolute risk difference is small, but statistically significant. “These findings do not necessarily warrant changing the management of treatment for individual patients,” wrote Annette Erlangsen, PhD, a researcher at the Danish Research Institute for Suicide Prevention in Hellerup, and colleagues. “As with all patients, physicians should be aware of the potential for depression, demoralization, and suicide.”

In addition, dementia, Alzheimer’s disease, and intellectual disabilities may be associated with lower suicide rates, according to the study, which was published in JAMA.

“Plausible mechanisms” could underlie the association between neurologic disease and suicide, the authors wrote. A neurologic diagnosis “may constitute a distressing life event,” and the diseases may have psychological, physical, and psychiatric effects. Patients may see themselves as a burden or have less financial security. In addition, the diseases may entail “communication difficulties, poor sleep, and pain.” Neurologic diseases may alter brain circuitry and functioning and influence aggression and impulsivity. “People with neurologic disorders may also have easier access to toxic medication,” they added.
 

More than a dozen conditions examined

Prior studies have found associations between neurologic conditions and rates of suicide, but data have been inconclusive or inconsistent for some of the disorders. To examine whether people with neurologic disorders have higher suicide rates, relative to people without these disorders, the researchers conducted a retrospective study. They analyzed data from more than 7.3 million people aged 15 years or older who lived in Denmark between 1980 and 2016. The cohort included more than 1.2 million people with neurologic disorders. The investigators identified neurologic disorders using ICD codes for head injury, stroke, epilepsy, polyneuropathy, diseases of the myoneural junction, Parkinson’s disease, multiple sclerosis, CNS infections, meningitis, encephalitis, amyotrophic lateral sclerosis, Huntington’s disease, dementia, intellectual disability, and other brain disorders. They compared incidence rates using a Poisson regression model and adjusted for time period, sex, age, region, socioeconomic status, comorbidity, self-harm or psychiatric hospitalization prior to a neurologic diagnosis, and whether a person lived alone.

In all, 35,483 people in the cohort died by suicide at an average age of about 52 years; 77.4% were male. About 15% of those who died by suicide had a neurologic disorder. The suicide incidence rate among people with a neurologic disorder was 44.0 per 100,000 person-years, whereas the rate among people without a neurologic disorder was 20.1 per 100,000 person-years.

The adjusted incidence rate ratio for people with a neurologic disorder was 1.8. The rate ratio was highest during the 3 months after diagnosis, at 3.1. Huntington’s disease and amyotrophic lateral sclerosis were associated with “the largest excess adjusted [incidence rate ratios] of suicide mortality,” with a rate ratio of 4.9 for each condition, the researchers reported. The adjusted incidence rate ratio was 1.7 for head injury, 1.3 for stroke, 1.7 for epilepsy, 1.4 for intracerebral hemorrhage, 1.3 for cerebral infarction, 1.3 for subarachnoid hemorrhage, 1.7 for polyneuropathy and peripheral neuropathy, 2.2 for Guillain-Barré syndrome, 1.9 for diseases of myoneural junction and muscle, 1.8 for other brain disorders, 1.7 for Parkinson’s disease, 2.2 for multiple sclerosis, and 1.6 for CNS infection.

Compared with people without a neurologic condition, people with dementia, Alzheimer’s disease, and intellectual disabilities had lower suicide rates, with adjusted incidence rate ratios of 0.8, 0.2, and 0.6, respectively. “However, the adjusted [incidence rate ratio] for people with dementia during the first month after diagnosis was 3.0,” the researchers wrote.

In addition, the suicide rate increased with an increasing cumulative number of hospital contacts for neurologic conditions.

 

 



Overall incidence rates declined

“Over the study period, the suicide incidence rate for people with neurological disorders decreased from 78.6 per 100,000 person-years during the 1980-1999 years to 27.3 per 100,000 person-years during the 2000-2016 years,” wrote Dr. Erlangsen and colleagues. “The suicide incidence rate for those without a disorder decreased from 26.3 to 12.7 during the same time spans. ... The decline in the overall suicide rate over time did not affect the relative risk pattern.”

The decline in the general suicide rate in Denmark “has largely been attributed to means restriction, such as efforts to limit availability of firearms and particularly toxic medication,” the authors added.

In those time spans, the adjusted incidence rate ratio for suicide among those with dementia decreased from 2.4 to 1.0, and among those with multiple sclerosis from 2.0 to 1.0. “It is possible that the improvements observed for dementia and multiple sclerosis may be related to improvements in treatment and intensified community-based support,” Dr. Erlangsen and coauthors wrote.

When the researchers used people with rheumatoid arthritis as a reference group, those with a neurologic disorder had a higher suicide rate per 100,000 person-years, 30.2 versus 18.4. The adjusted incidence rate ratio for that comparison was 1.4.

In patients with Huntington’s disease, depression mediated by hyperactivity in the hypothalamic-pituitary-adrenal axis may contribute to the risk of suicide. “Witnessing the course of the disease in one’s parent” also may contribute the risk, the researchers wrote.

The analysis may have missed people with neurologic disorders diagnosed before 1977 if they did not have subsequent contact with a hospital, the investigators noted. In addition, diagnoses given in primary care were not included, suicide deaths may be underrecorded, and “adjusting for preexisting mental disorders could be viewed as overadjusting,” they wrote.

The study was supported by a grant from the Psychiatric Research Foundation in Denmark. The authors reported that they had no disclosures.

SOURCE: Erlangsen A et al. JAMA. 2020 Feb 4. doi: 10.1001/jama.2019.21834.

People with neurologic disorders have a higher rate of suicide, compared with people without neurologic conditions, according to a population-based study in Denmark.

The absolute risk difference is small, but statistically significant. “These findings do not necessarily warrant changing the management of treatment for individual patients,” wrote Annette Erlangsen, PhD, a researcher at the Danish Research Institute for Suicide Prevention in Hellerup, and colleagues. “As with all patients, physicians should be aware of the potential for depression, demoralization, and suicide.”

In addition, dementia, Alzheimer’s disease, and intellectual disabilities may be associated with lower suicide rates, according to the study, which was published in JAMA.

“Plausible mechanisms” could underlie the association between neurologic disease and suicide, the authors wrote. A neurologic diagnosis “may constitute a distressing life event,” and the diseases may have psychological, physical, and psychiatric effects. Patients may see themselves as a burden or have less financial security. In addition, the diseases may entail “communication difficulties, poor sleep, and pain.” Neurologic diseases may alter brain circuitry and functioning and influence aggression and impulsivity. “People with neurologic disorders may also have easier access to toxic medication,” they added.
 

More than a dozen conditions examined

Prior studies have found associations between neurologic conditions and rates of suicide, but data have been inconclusive or inconsistent for some of the disorders. To examine whether people with neurologic disorders have higher suicide rates, relative to people without these disorders, the researchers conducted a retrospective study. They analyzed data from more than 7.3 million people aged 15 years or older who lived in Denmark between 1980 and 2016. The cohort included more than 1.2 million people with neurologic disorders. The investigators identified neurologic disorders using ICD codes for head injury, stroke, epilepsy, polyneuropathy, diseases of the myoneural junction, Parkinson’s disease, multiple sclerosis, CNS infections, meningitis, encephalitis, amyotrophic lateral sclerosis, Huntington’s disease, dementia, intellectual disability, and other brain disorders. They compared incidence rates using a Poisson regression model and adjusted for time period, sex, age, region, socioeconomic status, comorbidity, self-harm or psychiatric hospitalization prior to a neurologic diagnosis, and whether a person lived alone.

In all, 35,483 people in the cohort died by suicide at an average age of about 52 years; 77.4% were male. About 15% of those who died by suicide had a neurologic disorder. The suicide incidence rate among people with a neurologic disorder was 44.0 per 100,000 person-years, whereas the rate among people without a neurologic disorder was 20.1 per 100,000 person-years.

The adjusted incidence rate ratio for people with a neurologic disorder was 1.8. The rate ratio was highest during the 3 months after diagnosis, at 3.1. Huntington’s disease and amyotrophic lateral sclerosis were associated with “the largest excess adjusted [incidence rate ratios] of suicide mortality,” with a rate ratio of 4.9 for each condition, the researchers reported. The adjusted incidence rate ratio was 1.7 for head injury, 1.3 for stroke, 1.7 for epilepsy, 1.4 for intracerebral hemorrhage, 1.3 for cerebral infarction, 1.3 for subarachnoid hemorrhage, 1.7 for polyneuropathy and peripheral neuropathy, 2.2 for Guillain-Barré syndrome, 1.9 for diseases of myoneural junction and muscle, 1.8 for other brain disorders, 1.7 for Parkinson’s disease, 2.2 for multiple sclerosis, and 1.6 for CNS infection.

Compared with people without a neurologic condition, people with dementia, Alzheimer’s disease, and intellectual disabilities had lower suicide rates, with adjusted incidence rate ratios of 0.8, 0.2, and 0.6, respectively. “However, the adjusted [incidence rate ratio] for people with dementia during the first month after diagnosis was 3.0,” the researchers wrote.

In addition, the suicide rate increased with an increasing cumulative number of hospital contacts for neurologic conditions.

 

 



Overall incidence rates declined

“Over the study period, the suicide incidence rate for people with neurological disorders decreased from 78.6 per 100,000 person-years during the 1980-1999 years to 27.3 per 100,000 person-years during the 2000-2016 years,” wrote Dr. Erlangsen and colleagues. “The suicide incidence rate for those without a disorder decreased from 26.3 to 12.7 during the same time spans. ... The decline in the overall suicide rate over time did not affect the relative risk pattern.”

The decline in the general suicide rate in Denmark “has largely been attributed to means restriction, such as efforts to limit availability of firearms and particularly toxic medication,” the authors added.

In those time spans, the adjusted incidence rate ratio for suicide among those with dementia decreased from 2.4 to 1.0, and among those with multiple sclerosis from 2.0 to 1.0. “It is possible that the improvements observed for dementia and multiple sclerosis may be related to improvements in treatment and intensified community-based support,” Dr. Erlangsen and coauthors wrote.

When the researchers used people with rheumatoid arthritis as a reference group, those with a neurologic disorder had a higher suicide rate per 100,000 person-years, 30.2 versus 18.4. The adjusted incidence rate ratio for that comparison was 1.4.

In patients with Huntington’s disease, depression mediated by hyperactivity in the hypothalamic-pituitary-adrenal axis may contribute to the risk of suicide. “Witnessing the course of the disease in one’s parent” also may contribute the risk, the researchers wrote.

The analysis may have missed people with neurologic disorders diagnosed before 1977 if they did not have subsequent contact with a hospital, the investigators noted. In addition, diagnoses given in primary care were not included, suicide deaths may be underrecorded, and “adjusting for preexisting mental disorders could be viewed as overadjusting,” they wrote.

The study was supported by a grant from the Psychiatric Research Foundation in Denmark. The authors reported that they had no disclosures.

SOURCE: Erlangsen A et al. JAMA. 2020 Feb 4. doi: 10.1001/jama.2019.21834.

Issue
Neurology Reviews- 28(3)
Issue
Neurology Reviews- 28(3)
Publications
Publications
Topics
Article Type
Sections
Article Source

FROM JAMA

Citation Override
Publish date: February 4, 2020
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.

Losartan showing promise in pediatric epidermolysis bullosa trial

Article Type
Changed
Tue, 02/04/2020 - 12:01

– Treatment with the antihypertensive drug losartan raised no safety concerns in the treatment of children with epidermolysis bullosa (EB) and reduced signs of fibrosis in an early clinical study.

In the ongoing phase 1/2 REFLECT (Recessive dystrophic EB: Mechanisms of fibrosis and its prevention with Losartan in vivo) trial, involving 29 children, no severe complications have been noted so far, according to one of the study investigators, Dimitra Kiritsi, MD, of the University of Freiburg, Germany. At the EB World Congress, organized by the Dystrophic Epidermolysis Bullosa Association (DEBRA), she presented interim data on 18 patients in the trial, emphasizing that the primary aim of the trial was to evaluate the safety of this treatment approach.

Over the 2 years the trial has been underway, 65 adverse events have been reported, of which 4 have been severe. Two of these were bacterial infections that required hospital treatment and the other two were a reduction in the general health condition of the child.

Losartan is an angiotensin-II receptor blocker (ARB) that has been in clinical use for more than 25 years in adults and 15 years in children over the age of 6 years.

The drug may be used for treating recessive dystrophic EB (RDEB) in the future, Dr. Kiritsi said, because it attenuates tumor necrosis factor–beta (TGF-beta) signaling, which is thought to be involved in the fibrotic process. So while it may not target the genetic defect, it could help ameliorate the effects of the disease.

The precursor to REFLECT was a study performed in a mouse disease model of EB (EMBO Mol Med. 2015;7:1211-28) where a reduction in fibrotic scarring was seen with losartan with “remarkable effects” on “mitten” deformity, Dr. Kiritsi said. The results of that study suggested that the earlier treatment with losartan was started in the course of the disease, the better the effect, she added. (Mitten deformity is the result of fused skin between the fingers or toes, and the subsequent buildup of fibrotic tissue causes the hand or foot to contract.)

REFLECT is an investigator-initiated trial that started in 2017 and is being funded by DEBRA International. It is a dual-center, nonrandomized, single-arm study in which children aged 3-16 years with RDEB are treated with losartan for 10 months, with follow-up at 3 months.

Various secondary endpoints were included to look for the first signs of any efficacy: the Physician’s Global Assessment (PGA), the Birmingham Epidermolysis Bullosa Severity Score (BEBS), the Epidermolysis Bullosa Disease Activity and Scarring Index (EBDASI), the Itch Assessment Scale for the Pediatric Burn Patients, and two quality of life indices: the Quality of Life in EB (QOLEB) questionnaire and the Children’s Dermatology Life Quality Index (CDLQI).



Dr. Kiritsi highlighted a few of the secondary endpoint findings, saying that reduced BEBS scores showed there was “amelioration of the patients’ phenotype” and that EBDASI scores also decreased, with “nearly 60% of the patients having significant improvement of their skin disease.” Importantly, itch improved in most of the patients, she said. Reductions in CDLQI were observed, “meaning that quality of life was significantly better at the end of the trial.” There were also decreases in inflammatory markers, such as C-reactive protein, interleukin-6, and TNF-alpha.

Although there is no validated tool available to assess hand function, Dr. Kiritsi and her team used their own morphometric scoring instrument to measure how far the hand could stretch; their evaluations suggested that this measure improved – or at least did not worsen – with losartan treatment, she noted.

A larger, randomized trial is needed to confirm if there is any benefit of losartan, but first, a new, easy-to-swallow losartan formulation needs to be developed specifically for EB in the pediatric population, Dr. Kiritsi said. Although a pediatric suspension of losartan was previously available, it is no longer on the market, so the next step is to develop a formulation that could be used in a pivotal clinical trial, she noted.

“Losartan faces fewer technical hurdles compared to other novel treatments as it is an established medicine,” Dr. Kiritsi and associates observed in a poster presentation. There are still economic hurdles, however, since “with losartan patents expired, companies cannot expect to recoup an investment into clinical studies” and alternative funding sources are needed.

In 2019, losartan was granted an orphan drug designation for the treatment of EB from both the Food and Drug Administration and the European Medicines Agency, but its use remains off label in children. “We decided to treat children,” Dr. Kiritsi said, “because we wanted to start as early as possible. If you already have mitten deformities, these cannot be reversed.”

DEBRA International funded the study. Dr. Kiritsi received research support from Rheacell GmbH and honoraria or consultation fees from Amryt Pharma and Rheacell GmbH. She has received other support from DEBRA International, EB Research Partnership, Fritz Thyssen Stiftung, German Research Foundation (funding of research projects), and 3R Pharma Consulting and Midas Pharma GmbH (consultation for losartan new drug formulation).

SOURCE: Kiritsi D et al. EB 2020. Poster 47.
 

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

– Treatment with the antihypertensive drug losartan raised no safety concerns in the treatment of children with epidermolysis bullosa (EB) and reduced signs of fibrosis in an early clinical study.

In the ongoing phase 1/2 REFLECT (Recessive dystrophic EB: Mechanisms of fibrosis and its prevention with Losartan in vivo) trial, involving 29 children, no severe complications have been noted so far, according to one of the study investigators, Dimitra Kiritsi, MD, of the University of Freiburg, Germany. At the EB World Congress, organized by the Dystrophic Epidermolysis Bullosa Association (DEBRA), she presented interim data on 18 patients in the trial, emphasizing that the primary aim of the trial was to evaluate the safety of this treatment approach.

Over the 2 years the trial has been underway, 65 adverse events have been reported, of which 4 have been severe. Two of these were bacterial infections that required hospital treatment and the other two were a reduction in the general health condition of the child.

Losartan is an angiotensin-II receptor blocker (ARB) that has been in clinical use for more than 25 years in adults and 15 years in children over the age of 6 years.

The drug may be used for treating recessive dystrophic EB (RDEB) in the future, Dr. Kiritsi said, because it attenuates tumor necrosis factor–beta (TGF-beta) signaling, which is thought to be involved in the fibrotic process. So while it may not target the genetic defect, it could help ameliorate the effects of the disease.

The precursor to REFLECT was a study performed in a mouse disease model of EB (EMBO Mol Med. 2015;7:1211-28) where a reduction in fibrotic scarring was seen with losartan with “remarkable effects” on “mitten” deformity, Dr. Kiritsi said. The results of that study suggested that the earlier treatment with losartan was started in the course of the disease, the better the effect, she added. (Mitten deformity is the result of fused skin between the fingers or toes, and the subsequent buildup of fibrotic tissue causes the hand or foot to contract.)

REFLECT is an investigator-initiated trial that started in 2017 and is being funded by DEBRA International. It is a dual-center, nonrandomized, single-arm study in which children aged 3-16 years with RDEB are treated with losartan for 10 months, with follow-up at 3 months.

Various secondary endpoints were included to look for the first signs of any efficacy: the Physician’s Global Assessment (PGA), the Birmingham Epidermolysis Bullosa Severity Score (BEBS), the Epidermolysis Bullosa Disease Activity and Scarring Index (EBDASI), the Itch Assessment Scale for the Pediatric Burn Patients, and two quality of life indices: the Quality of Life in EB (QOLEB) questionnaire and the Children’s Dermatology Life Quality Index (CDLQI).



Dr. Kiritsi highlighted a few of the secondary endpoint findings, saying that reduced BEBS scores showed there was “amelioration of the patients’ phenotype” and that EBDASI scores also decreased, with “nearly 60% of the patients having significant improvement of their skin disease.” Importantly, itch improved in most of the patients, she said. Reductions in CDLQI were observed, “meaning that quality of life was significantly better at the end of the trial.” There were also decreases in inflammatory markers, such as C-reactive protein, interleukin-6, and TNF-alpha.

Although there is no validated tool available to assess hand function, Dr. Kiritsi and her team used their own morphometric scoring instrument to measure how far the hand could stretch; their evaluations suggested that this measure improved – or at least did not worsen – with losartan treatment, she noted.

A larger, randomized trial is needed to confirm if there is any benefit of losartan, but first, a new, easy-to-swallow losartan formulation needs to be developed specifically for EB in the pediatric population, Dr. Kiritsi said. Although a pediatric suspension of losartan was previously available, it is no longer on the market, so the next step is to develop a formulation that could be used in a pivotal clinical trial, she noted.

“Losartan faces fewer technical hurdles compared to other novel treatments as it is an established medicine,” Dr. Kiritsi and associates observed in a poster presentation. There are still economic hurdles, however, since “with losartan patents expired, companies cannot expect to recoup an investment into clinical studies” and alternative funding sources are needed.

In 2019, losartan was granted an orphan drug designation for the treatment of EB from both the Food and Drug Administration and the European Medicines Agency, but its use remains off label in children. “We decided to treat children,” Dr. Kiritsi said, “because we wanted to start as early as possible. If you already have mitten deformities, these cannot be reversed.”

DEBRA International funded the study. Dr. Kiritsi received research support from Rheacell GmbH and honoraria or consultation fees from Amryt Pharma and Rheacell GmbH. She has received other support from DEBRA International, EB Research Partnership, Fritz Thyssen Stiftung, German Research Foundation (funding of research projects), and 3R Pharma Consulting and Midas Pharma GmbH (consultation for losartan new drug formulation).

SOURCE: Kiritsi D et al. EB 2020. Poster 47.
 

– Treatment with the antihypertensive drug losartan raised no safety concerns in the treatment of children with epidermolysis bullosa (EB) and reduced signs of fibrosis in an early clinical study.

In the ongoing phase 1/2 REFLECT (Recessive dystrophic EB: Mechanisms of fibrosis and its prevention with Losartan in vivo) trial, involving 29 children, no severe complications have been noted so far, according to one of the study investigators, Dimitra Kiritsi, MD, of the University of Freiburg, Germany. At the EB World Congress, organized by the Dystrophic Epidermolysis Bullosa Association (DEBRA), she presented interim data on 18 patients in the trial, emphasizing that the primary aim of the trial was to evaluate the safety of this treatment approach.

Over the 2 years the trial has been underway, 65 adverse events have been reported, of which 4 have been severe. Two of these were bacterial infections that required hospital treatment and the other two were a reduction in the general health condition of the child.

Losartan is an angiotensin-II receptor blocker (ARB) that has been in clinical use for more than 25 years in adults and 15 years in children over the age of 6 years.

The drug may be used for treating recessive dystrophic EB (RDEB) in the future, Dr. Kiritsi said, because it attenuates tumor necrosis factor–beta (TGF-beta) signaling, which is thought to be involved in the fibrotic process. So while it may not target the genetic defect, it could help ameliorate the effects of the disease.

The precursor to REFLECT was a study performed in a mouse disease model of EB (EMBO Mol Med. 2015;7:1211-28) where a reduction in fibrotic scarring was seen with losartan with “remarkable effects” on “mitten” deformity, Dr. Kiritsi said. The results of that study suggested that the earlier treatment with losartan was started in the course of the disease, the better the effect, she added. (Mitten deformity is the result of fused skin between the fingers or toes, and the subsequent buildup of fibrotic tissue causes the hand or foot to contract.)

REFLECT is an investigator-initiated trial that started in 2017 and is being funded by DEBRA International. It is a dual-center, nonrandomized, single-arm study in which children aged 3-16 years with RDEB are treated with losartan for 10 months, with follow-up at 3 months.

Various secondary endpoints were included to look for the first signs of any efficacy: the Physician’s Global Assessment (PGA), the Birmingham Epidermolysis Bullosa Severity Score (BEBS), the Epidermolysis Bullosa Disease Activity and Scarring Index (EBDASI), the Itch Assessment Scale for the Pediatric Burn Patients, and two quality of life indices: the Quality of Life in EB (QOLEB) questionnaire and the Children’s Dermatology Life Quality Index (CDLQI).



Dr. Kiritsi highlighted a few of the secondary endpoint findings, saying that reduced BEBS scores showed there was “amelioration of the patients’ phenotype” and that EBDASI scores also decreased, with “nearly 60% of the patients having significant improvement of their skin disease.” Importantly, itch improved in most of the patients, she said. Reductions in CDLQI were observed, “meaning that quality of life was significantly better at the end of the trial.” There were also decreases in inflammatory markers, such as C-reactive protein, interleukin-6, and TNF-alpha.

Although there is no validated tool available to assess hand function, Dr. Kiritsi and her team used their own morphometric scoring instrument to measure how far the hand could stretch; their evaluations suggested that this measure improved – or at least did not worsen – with losartan treatment, she noted.

A larger, randomized trial is needed to confirm if there is any benefit of losartan, but first, a new, easy-to-swallow losartan formulation needs to be developed specifically for EB in the pediatric population, Dr. Kiritsi said. Although a pediatric suspension of losartan was previously available, it is no longer on the market, so the next step is to develop a formulation that could be used in a pivotal clinical trial, she noted.

“Losartan faces fewer technical hurdles compared to other novel treatments as it is an established medicine,” Dr. Kiritsi and associates observed in a poster presentation. There are still economic hurdles, however, since “with losartan patents expired, companies cannot expect to recoup an investment into clinical studies” and alternative funding sources are needed.

In 2019, losartan was granted an orphan drug designation for the treatment of EB from both the Food and Drug Administration and the European Medicines Agency, but its use remains off label in children. “We decided to treat children,” Dr. Kiritsi said, “because we wanted to start as early as possible. If you already have mitten deformities, these cannot be reversed.”

DEBRA International funded the study. Dr. Kiritsi received research support from Rheacell GmbH and honoraria or consultation fees from Amryt Pharma and Rheacell GmbH. She has received other support from DEBRA International, EB Research Partnership, Fritz Thyssen Stiftung, German Research Foundation (funding of research projects), and 3R Pharma Consulting and Midas Pharma GmbH (consultation for losartan new drug formulation).

SOURCE: Kiritsi D et al. EB 2020. Poster 47.
 

Publications
Publications
Topics
Article Type
Sections
Article Source

REPORTING FROM EB 2020

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.

Distinct Violaceous Plaques in Conjunction With Blisters

Article Type
Changed
Wed, 02/05/2020 - 11:03
Display Headline
Distinct Violaceous Plaques in Conjunction With Blisters

The Diagnosis: Lichen Planus Pemphigoides 

Lichen planus pemphigoides (LPP) is a rare autoimmune subepithelial blistering disorder with clinical, pathologic, and immunologic features of lichen planus (LP) and bullous pemphigoid (BP).1 It mainly arises in adults and usually is idiopathic but has been associated with certain infections,2 drugs such as angiotensin-converting enzyme inhibitors,3 phototherapy,4 and malignancy.5 Patients classically present with lichenoid lesions, tense vesiculobullae, and erosions.6 Vesiculobullae formation usually follows the development of lichenoid lesions, occurs on both lichenoid lesions and unaffected skin, and predominantly involves the lower extremities, as in our patient.1,6 

The pathogenesis of LPP is not fully understood but likely represents a distinct entity rather than a subtype of BP or the simultaneous occurrence of LP and BP. Lichen planus pemphigoides generally has an earlier onset and better treatment response compared to BP.7 Further, autoantibodies in patients with LPP react to a novel epitope within the C-terminal portion of the BP-180 NC16A domain. Accordingly, it has been postulated that an inflammatory cutaneous process resulting from infection, phototherapy, or LP itself leads to damage of the epidermis and triggers a secondary blistering autoimmune dermatosis mediated by antibody formation against basement membrane (BM) antigens, such as BP-180.

The diagnosis of LPP ultimately is confirmed with immunohistologic analysis. Biopsy of LPP shows findings consistent with both LP and BP (quiz image [top]). In the lichenoid portion, biopsy reveals orthohyperkeratosis, hypergranulosis, and acanthosis of the epidermis; a bandlike infiltrate consisting primarily of lymphocytes in the upper dermis; and apoptotic keratinocytes (colloid bodies) and vacuolar degeneration at the dermoepidermal junction (DEJ).1 Biopsy of bullae reveals eosinophilic spongiosis, a subepithelial blister plane with eosinophils, and a mixed superficial inflammatory cell infiltrate. Direct immunofluorescence from perilesional skin reveals linear deposition of IgG and/or C3 at the DEJ (quiz image [bottom]).1 Measurement of anti-BM antibodies against BP-180 and BP-230 can be useful in suspected cases, as 50% to 60% of patients have circulating antibodies against these antigens.6 Remission usually is achieved with topical and systemic corticosteroids and/or steroid-sparing agents, with rare recurrence following lesion resolution.1 More recently, successful treatment with biologics such as ustekinumab has been reported.8 

The predominant differential diagnosis for LPP is bullous LP, a variant of LP in which vesiculobullous disease occurs exclusively on preexisting LP lesions, commonly on the legs due to severe vacuolar degeneration at the DEJ. On histopathology, the characteristic features of LP (eg, orthohyperkeratosis, hypergranulosis, acanthosis, bandlike lymphocytic infiltrate, colloid bodies) along with subepidermal clefting will be seen. However, in bullous LP (Figure 1) there is an absence of linear IgG and/or C3 deposition at the DEJ on direct immunofluorescence. Furthermore, patients lack circulating antibodies against BP-180 and BP-230.9 

Figure 1. Bullous lichen planus. Bandlike inflammatory infiltrate at the level of the dermoepidermal junction composed of lymphocytes and histiocytes with melanin pigment incontinence. There is a sawtooth pattern of the rete ridges and subepidermal clefting (Max-Joseph space)(H&E, original magnification ×200).

Lichen planus pemphigoides also can be confused with BP. Bullous pemphigoid is the most common autoimmune blistering disorder; typically arises in older adults; and is caused by autoantibody formation against hemidesmosomal proteins, particularly BP-180 and BP-230. Patients classically present with tense bullae and erosions on an erythematous, urticarial, or normal base. These lesions often are pruritic and concentrated on the trunk, axillary and inguinal folds, and extremity flexures. Histopathologic examination of a bulla edge reveals the classic findings seen in BP (eg, eosinophilic spongiosis, subepithelial blister plane with eosinophils)(Figure 2). Direct immunofluorescence of perilesional skin reveals linear IgG and/or C3 deposition along the DEJ. A large subset of patients also has circulating antibodies against BP-180 and BP-230. In contrast to LPP, however, patients with BP do not develop lichenoid lesions clinically or a lichenoid tissue reaction histopathologically.10 

Figure 2. Bullous pemphigoid. Subepidermal vesicle containing numerous eosinophils. A moderately intense eosinophilic infiltrate is present in the dermis (H&E, original magnification ×200).

Bullous systemic lupus erythematosus (SLE), a rare cutaneous manifestation of SLE, typically arises in young women of African descent and is due to autoantibody formation against type VII collagen and other BM-zone antigens. Patients generally present with acute onset of tense vesiculobullae on a normal or erythematous base, which often are transient and heal without milia or scarring. Common sites of involvement include the trunk, arms, neck, face, and vermilion border, as well as the oral mucosa. The diagnosis of bullous SLE requires that patients fulfill the criteria for SLE and is confirmed by immunohistologic analysis. Biopsy of a bulla edge reveals a subepidermal blister containing neutrophils and increased mucin within the reticular dermis (Figure 3). Direct immunofluorescence of perilesional skin most commonly reveals linear and/or granular deposition of IgG, IgA, C3, and IgM at the DEJ.11 

Figure 3. Bullous systemic lupus erythematosus. Subepidermal vesicle with numerous neutrophils. Within the dermis there is a moderately intense neutrophilic inflammation. Necrosis of the keratinocytes is present in an adjacent follicle (H&E, original magnification ×200).

Bullous tinea is a manifestation of cutaneous dermatophytosis that usually occurs in the setting of tinea pedis. Common causative dermatophytes include Trichophyton mentagrophytes, Trichophyton rubrum, and Epidermophyton floccosum. Diagnosis is made by demonstration of fungal hyphae on potassium hydroxide preparation of the blister roof, biopsy with periodic acid-Schiff stain, or fungal culture. If routine histopathologic analysis is performed, epidermal spongiosis with varying degrees of papillary dermal edema is seen, along with abundant fungal elements in the stratum corneum (Figure 4). Direct immunofluorescence of perilesional skin usually is negative, but C3 deposition in a linear and/or granular pattern along the DEJ has been reported.12 

Figure 4. Bullous tinea. Subcorneal pustule with numerous neutrophils and eosinophils. Notable numbers of eosinophils and neutrophils are present in the dermis with extension into the epidermis (H&E, original magnification ×200). Fungal hyphae consistent with Tinea species were present within the stratum corneum (Periodic acid–Schiff, original magnification ×600 [inset]). 

Lichen planus pemphigoides is a rare disease entity and often presents a diagnostic challenge to clinicians. The differential for LPP includes bullous LP as well as other bullous disorders. Ultimately, the diagnosis is confirmed through immunohistologic analysis. Timely diagnosis of LPP is crucial, as most patients can achieve long-term remission with appropriate treatment. 

References
  1. Zaraa I, Mahfoudh A, Sellami MK, et al. Lichen planus pemphigoides: four new cases and a review of the literature. Int J Dermatol. 2013;52:406-412. 
  2. Mohanarao TS, Kumar GA, Chennamsetty K, et al. Childhood lichen planus pemphigoides triggered by chickenpox. Indian Dermatol Online J. 2014;5:S98-S100. 
  3. Onprasert W, Chanprapaph K. Lichen planus pemphigoides induced by enalapril: a case report and a review of literature. Case Rep Dermatol. 2017;9:217-224. 
  4. Kuramoto N, Kishimoto S, Shibagaki R, et al. PUVA-induced lichen planus pemphigoides. Br J Dermatol. 2000;142:509-512. 
  5. Shimada H, Shono T, Sakai T, et al. Lichen planus pemphigoides concomitant with rectal adenocarcinoma: fortuitous or a true association? Eur J Dermatol. 2015;25:501-503. 
  6. Matos-Pires E, Campos S, Lencastre A, et al. Lichen planus pemphigoides. J Dtsch Dermatol Ges. 2018;16:335-337. 
  7. Zillikens D, Caux F, Mascaro JM, et al. Autoantibodies in lichen planus pemphigoides react with a novel epitope within the C-terminal NC16A domain of BP180. J Invest Dermatol. 1999;113:117-121. 
  8. Knisley RR, Petropolis AA, Mackey VT. Lichen planus pemphigoides treated with ustekinumab. Cutis. 2017;100:415-418. 
  9. Wagner G, Rose C, Sachse MM. Clinical variants of lichen planus. J Dtsch Dermatol Ges. 2013;11:309-319. 
  10. Bagci IS, Horvath ON, Ruzicka T, et al. Bullous pemphigoid. Autoimmun Rev. 2017;16:445-455. 
  11. Contestable JJ, Edhegard KD, Meyerle JH. Bullous systemic lupus erythematosus: a review and update to diagnosis and treatment. Am J Clin Dermatol. 2014;15:517-524. 
  12. Miller DD, Bhawan J. Bullous tinea pedis with direct immunofluorescence positivity: when is a positive result not autoimmune bullous disease? Am J Dermatopathol. 2013;35:587-594.
Article PDF
Author and Disclosure Information

Drs. Grandhi, Tjarks, Lin, Ferringer, and Hossler are from the Department of Dermatology, Geisinger Medical Center, Danville, Pennsylvania. Dr. Bazewicz is from the College of Medicine, Penn State Hershey Medical Center.

The authors report no conflict of interest.

Correspondence: Radhika Grandhi, MD, MPH, Department of Dermatology, Geisinger Medical Center, 115 Woodbine Ln, Danville, PA 17822 (rrgrandhi@geisinger.edu).

Issue
Cutis - 105(2)
Publications
Topics
Page Number
62, 71-72
Sections
Author and Disclosure Information

Drs. Grandhi, Tjarks, Lin, Ferringer, and Hossler are from the Department of Dermatology, Geisinger Medical Center, Danville, Pennsylvania. Dr. Bazewicz is from the College of Medicine, Penn State Hershey Medical Center.

The authors report no conflict of interest.

Correspondence: Radhika Grandhi, MD, MPH, Department of Dermatology, Geisinger Medical Center, 115 Woodbine Ln, Danville, PA 17822 (rrgrandhi@geisinger.edu).

Author and Disclosure Information

Drs. Grandhi, Tjarks, Lin, Ferringer, and Hossler are from the Department of Dermatology, Geisinger Medical Center, Danville, Pennsylvania. Dr. Bazewicz is from the College of Medicine, Penn State Hershey Medical Center.

The authors report no conflict of interest.

Correspondence: Radhika Grandhi, MD, MPH, Department of Dermatology, Geisinger Medical Center, 115 Woodbine Ln, Danville, PA 17822 (rrgrandhi@geisinger.edu).

Article PDF
Article PDF
Related Articles

The Diagnosis: Lichen Planus Pemphigoides 

Lichen planus pemphigoides (LPP) is a rare autoimmune subepithelial blistering disorder with clinical, pathologic, and immunologic features of lichen planus (LP) and bullous pemphigoid (BP).1 It mainly arises in adults and usually is idiopathic but has been associated with certain infections,2 drugs such as angiotensin-converting enzyme inhibitors,3 phototherapy,4 and malignancy.5 Patients classically present with lichenoid lesions, tense vesiculobullae, and erosions.6 Vesiculobullae formation usually follows the development of lichenoid lesions, occurs on both lichenoid lesions and unaffected skin, and predominantly involves the lower extremities, as in our patient.1,6 

The pathogenesis of LPP is not fully understood but likely represents a distinct entity rather than a subtype of BP or the simultaneous occurrence of LP and BP. Lichen planus pemphigoides generally has an earlier onset and better treatment response compared to BP.7 Further, autoantibodies in patients with LPP react to a novel epitope within the C-terminal portion of the BP-180 NC16A domain. Accordingly, it has been postulated that an inflammatory cutaneous process resulting from infection, phototherapy, or LP itself leads to damage of the epidermis and triggers a secondary blistering autoimmune dermatosis mediated by antibody formation against basement membrane (BM) antigens, such as BP-180.

The diagnosis of LPP ultimately is confirmed with immunohistologic analysis. Biopsy of LPP shows findings consistent with both LP and BP (quiz image [top]). In the lichenoid portion, biopsy reveals orthohyperkeratosis, hypergranulosis, and acanthosis of the epidermis; a bandlike infiltrate consisting primarily of lymphocytes in the upper dermis; and apoptotic keratinocytes (colloid bodies) and vacuolar degeneration at the dermoepidermal junction (DEJ).1 Biopsy of bullae reveals eosinophilic spongiosis, a subepithelial blister plane with eosinophils, and a mixed superficial inflammatory cell infiltrate. Direct immunofluorescence from perilesional skin reveals linear deposition of IgG and/or C3 at the DEJ (quiz image [bottom]).1 Measurement of anti-BM antibodies against BP-180 and BP-230 can be useful in suspected cases, as 50% to 60% of patients have circulating antibodies against these antigens.6 Remission usually is achieved with topical and systemic corticosteroids and/or steroid-sparing agents, with rare recurrence following lesion resolution.1 More recently, successful treatment with biologics such as ustekinumab has been reported.8 

The predominant differential diagnosis for LPP is bullous LP, a variant of LP in which vesiculobullous disease occurs exclusively on preexisting LP lesions, commonly on the legs due to severe vacuolar degeneration at the DEJ. On histopathology, the characteristic features of LP (eg, orthohyperkeratosis, hypergranulosis, acanthosis, bandlike lymphocytic infiltrate, colloid bodies) along with subepidermal clefting will be seen. However, in bullous LP (Figure 1) there is an absence of linear IgG and/or C3 deposition at the DEJ on direct immunofluorescence. Furthermore, patients lack circulating antibodies against BP-180 and BP-230.9 

Figure 1. Bullous lichen planus. Bandlike inflammatory infiltrate at the level of the dermoepidermal junction composed of lymphocytes and histiocytes with melanin pigment incontinence. There is a sawtooth pattern of the rete ridges and subepidermal clefting (Max-Joseph space)(H&E, original magnification ×200).

Lichen planus pemphigoides also can be confused with BP. Bullous pemphigoid is the most common autoimmune blistering disorder; typically arises in older adults; and is caused by autoantibody formation against hemidesmosomal proteins, particularly BP-180 and BP-230. Patients classically present with tense bullae and erosions on an erythematous, urticarial, or normal base. These lesions often are pruritic and concentrated on the trunk, axillary and inguinal folds, and extremity flexures. Histopathologic examination of a bulla edge reveals the classic findings seen in BP (eg, eosinophilic spongiosis, subepithelial blister plane with eosinophils)(Figure 2). Direct immunofluorescence of perilesional skin reveals linear IgG and/or C3 deposition along the DEJ. A large subset of patients also has circulating antibodies against BP-180 and BP-230. In contrast to LPP, however, patients with BP do not develop lichenoid lesions clinically or a lichenoid tissue reaction histopathologically.10 

Figure 2. Bullous pemphigoid. Subepidermal vesicle containing numerous eosinophils. A moderately intense eosinophilic infiltrate is present in the dermis (H&E, original magnification ×200).

Bullous systemic lupus erythematosus (SLE), a rare cutaneous manifestation of SLE, typically arises in young women of African descent and is due to autoantibody formation against type VII collagen and other BM-zone antigens. Patients generally present with acute onset of tense vesiculobullae on a normal or erythematous base, which often are transient and heal without milia or scarring. Common sites of involvement include the trunk, arms, neck, face, and vermilion border, as well as the oral mucosa. The diagnosis of bullous SLE requires that patients fulfill the criteria for SLE and is confirmed by immunohistologic analysis. Biopsy of a bulla edge reveals a subepidermal blister containing neutrophils and increased mucin within the reticular dermis (Figure 3). Direct immunofluorescence of perilesional skin most commonly reveals linear and/or granular deposition of IgG, IgA, C3, and IgM at the DEJ.11 

Figure 3. Bullous systemic lupus erythematosus. Subepidermal vesicle with numerous neutrophils. Within the dermis there is a moderately intense neutrophilic inflammation. Necrosis of the keratinocytes is present in an adjacent follicle (H&E, original magnification ×200).

Bullous tinea is a manifestation of cutaneous dermatophytosis that usually occurs in the setting of tinea pedis. Common causative dermatophytes include Trichophyton mentagrophytes, Trichophyton rubrum, and Epidermophyton floccosum. Diagnosis is made by demonstration of fungal hyphae on potassium hydroxide preparation of the blister roof, biopsy with periodic acid-Schiff stain, or fungal culture. If routine histopathologic analysis is performed, epidermal spongiosis with varying degrees of papillary dermal edema is seen, along with abundant fungal elements in the stratum corneum (Figure 4). Direct immunofluorescence of perilesional skin usually is negative, but C3 deposition in a linear and/or granular pattern along the DEJ has been reported.12 

Figure 4. Bullous tinea. Subcorneal pustule with numerous neutrophils and eosinophils. Notable numbers of eosinophils and neutrophils are present in the dermis with extension into the epidermis (H&E, original magnification ×200). Fungal hyphae consistent with Tinea species were present within the stratum corneum (Periodic acid–Schiff, original magnification ×600 [inset]). 

Lichen planus pemphigoides is a rare disease entity and often presents a diagnostic challenge to clinicians. The differential for LPP includes bullous LP as well as other bullous disorders. Ultimately, the diagnosis is confirmed through immunohistologic analysis. Timely diagnosis of LPP is crucial, as most patients can achieve long-term remission with appropriate treatment. 

The Diagnosis: Lichen Planus Pemphigoides 

Lichen planus pemphigoides (LPP) is a rare autoimmune subepithelial blistering disorder with clinical, pathologic, and immunologic features of lichen planus (LP) and bullous pemphigoid (BP).1 It mainly arises in adults and usually is idiopathic but has been associated with certain infections,2 drugs such as angiotensin-converting enzyme inhibitors,3 phototherapy,4 and malignancy.5 Patients classically present with lichenoid lesions, tense vesiculobullae, and erosions.6 Vesiculobullae formation usually follows the development of lichenoid lesions, occurs on both lichenoid lesions and unaffected skin, and predominantly involves the lower extremities, as in our patient.1,6 

The pathogenesis of LPP is not fully understood but likely represents a distinct entity rather than a subtype of BP or the simultaneous occurrence of LP and BP. Lichen planus pemphigoides generally has an earlier onset and better treatment response compared to BP.7 Further, autoantibodies in patients with LPP react to a novel epitope within the C-terminal portion of the BP-180 NC16A domain. Accordingly, it has been postulated that an inflammatory cutaneous process resulting from infection, phototherapy, or LP itself leads to damage of the epidermis and triggers a secondary blistering autoimmune dermatosis mediated by antibody formation against basement membrane (BM) antigens, such as BP-180.

The diagnosis of LPP ultimately is confirmed with immunohistologic analysis. Biopsy of LPP shows findings consistent with both LP and BP (quiz image [top]). In the lichenoid portion, biopsy reveals orthohyperkeratosis, hypergranulosis, and acanthosis of the epidermis; a bandlike infiltrate consisting primarily of lymphocytes in the upper dermis; and apoptotic keratinocytes (colloid bodies) and vacuolar degeneration at the dermoepidermal junction (DEJ).1 Biopsy of bullae reveals eosinophilic spongiosis, a subepithelial blister plane with eosinophils, and a mixed superficial inflammatory cell infiltrate. Direct immunofluorescence from perilesional skin reveals linear deposition of IgG and/or C3 at the DEJ (quiz image [bottom]).1 Measurement of anti-BM antibodies against BP-180 and BP-230 can be useful in suspected cases, as 50% to 60% of patients have circulating antibodies against these antigens.6 Remission usually is achieved with topical and systemic corticosteroids and/or steroid-sparing agents, with rare recurrence following lesion resolution.1 More recently, successful treatment with biologics such as ustekinumab has been reported.8 

The predominant differential diagnosis for LPP is bullous LP, a variant of LP in which vesiculobullous disease occurs exclusively on preexisting LP lesions, commonly on the legs due to severe vacuolar degeneration at the DEJ. On histopathology, the characteristic features of LP (eg, orthohyperkeratosis, hypergranulosis, acanthosis, bandlike lymphocytic infiltrate, colloid bodies) along with subepidermal clefting will be seen. However, in bullous LP (Figure 1) there is an absence of linear IgG and/or C3 deposition at the DEJ on direct immunofluorescence. Furthermore, patients lack circulating antibodies against BP-180 and BP-230.9 

Figure 1. Bullous lichen planus. Bandlike inflammatory infiltrate at the level of the dermoepidermal junction composed of lymphocytes and histiocytes with melanin pigment incontinence. There is a sawtooth pattern of the rete ridges and subepidermal clefting (Max-Joseph space)(H&E, original magnification ×200).

Lichen planus pemphigoides also can be confused with BP. Bullous pemphigoid is the most common autoimmune blistering disorder; typically arises in older adults; and is caused by autoantibody formation against hemidesmosomal proteins, particularly BP-180 and BP-230. Patients classically present with tense bullae and erosions on an erythematous, urticarial, or normal base. These lesions often are pruritic and concentrated on the trunk, axillary and inguinal folds, and extremity flexures. Histopathologic examination of a bulla edge reveals the classic findings seen in BP (eg, eosinophilic spongiosis, subepithelial blister plane with eosinophils)(Figure 2). Direct immunofluorescence of perilesional skin reveals linear IgG and/or C3 deposition along the DEJ. A large subset of patients also has circulating antibodies against BP-180 and BP-230. In contrast to LPP, however, patients with BP do not develop lichenoid lesions clinically or a lichenoid tissue reaction histopathologically.10 

Figure 2. Bullous pemphigoid. Subepidermal vesicle containing numerous eosinophils. A moderately intense eosinophilic infiltrate is present in the dermis (H&E, original magnification ×200).

Bullous systemic lupus erythematosus (SLE), a rare cutaneous manifestation of SLE, typically arises in young women of African descent and is due to autoantibody formation against type VII collagen and other BM-zone antigens. Patients generally present with acute onset of tense vesiculobullae on a normal or erythematous base, which often are transient and heal without milia or scarring. Common sites of involvement include the trunk, arms, neck, face, and vermilion border, as well as the oral mucosa. The diagnosis of bullous SLE requires that patients fulfill the criteria for SLE and is confirmed by immunohistologic analysis. Biopsy of a bulla edge reveals a subepidermal blister containing neutrophils and increased mucin within the reticular dermis (Figure 3). Direct immunofluorescence of perilesional skin most commonly reveals linear and/or granular deposition of IgG, IgA, C3, and IgM at the DEJ.11 

Figure 3. Bullous systemic lupus erythematosus. Subepidermal vesicle with numerous neutrophils. Within the dermis there is a moderately intense neutrophilic inflammation. Necrosis of the keratinocytes is present in an adjacent follicle (H&E, original magnification ×200).

Bullous tinea is a manifestation of cutaneous dermatophytosis that usually occurs in the setting of tinea pedis. Common causative dermatophytes include Trichophyton mentagrophytes, Trichophyton rubrum, and Epidermophyton floccosum. Diagnosis is made by demonstration of fungal hyphae on potassium hydroxide preparation of the blister roof, biopsy with periodic acid-Schiff stain, or fungal culture. If routine histopathologic analysis is performed, epidermal spongiosis with varying degrees of papillary dermal edema is seen, along with abundant fungal elements in the stratum corneum (Figure 4). Direct immunofluorescence of perilesional skin usually is negative, but C3 deposition in a linear and/or granular pattern along the DEJ has been reported.12 

Figure 4. Bullous tinea. Subcorneal pustule with numerous neutrophils and eosinophils. Notable numbers of eosinophils and neutrophils are present in the dermis with extension into the epidermis (H&E, original magnification ×200). Fungal hyphae consistent with Tinea species were present within the stratum corneum (Periodic acid–Schiff, original magnification ×600 [inset]). 

Lichen planus pemphigoides is a rare disease entity and often presents a diagnostic challenge to clinicians. The differential for LPP includes bullous LP as well as other bullous disorders. Ultimately, the diagnosis is confirmed through immunohistologic analysis. Timely diagnosis of LPP is crucial, as most patients can achieve long-term remission with appropriate treatment. 

References
  1. Zaraa I, Mahfoudh A, Sellami MK, et al. Lichen planus pemphigoides: four new cases and a review of the literature. Int J Dermatol. 2013;52:406-412. 
  2. Mohanarao TS, Kumar GA, Chennamsetty K, et al. Childhood lichen planus pemphigoides triggered by chickenpox. Indian Dermatol Online J. 2014;5:S98-S100. 
  3. Onprasert W, Chanprapaph K. Lichen planus pemphigoides induced by enalapril: a case report and a review of literature. Case Rep Dermatol. 2017;9:217-224. 
  4. Kuramoto N, Kishimoto S, Shibagaki R, et al. PUVA-induced lichen planus pemphigoides. Br J Dermatol. 2000;142:509-512. 
  5. Shimada H, Shono T, Sakai T, et al. Lichen planus pemphigoides concomitant with rectal adenocarcinoma: fortuitous or a true association? Eur J Dermatol. 2015;25:501-503. 
  6. Matos-Pires E, Campos S, Lencastre A, et al. Lichen planus pemphigoides. J Dtsch Dermatol Ges. 2018;16:335-337. 
  7. Zillikens D, Caux F, Mascaro JM, et al. Autoantibodies in lichen planus pemphigoides react with a novel epitope within the C-terminal NC16A domain of BP180. J Invest Dermatol. 1999;113:117-121. 
  8. Knisley RR, Petropolis AA, Mackey VT. Lichen planus pemphigoides treated with ustekinumab. Cutis. 2017;100:415-418. 
  9. Wagner G, Rose C, Sachse MM. Clinical variants of lichen planus. J Dtsch Dermatol Ges. 2013;11:309-319. 
  10. Bagci IS, Horvath ON, Ruzicka T, et al. Bullous pemphigoid. Autoimmun Rev. 2017;16:445-455. 
  11. Contestable JJ, Edhegard KD, Meyerle JH. Bullous systemic lupus erythematosus: a review and update to diagnosis and treatment. Am J Clin Dermatol. 2014;15:517-524. 
  12. Miller DD, Bhawan J. Bullous tinea pedis with direct immunofluorescence positivity: when is a positive result not autoimmune bullous disease? Am J Dermatopathol. 2013;35:587-594.
References
  1. Zaraa I, Mahfoudh A, Sellami MK, et al. Lichen planus pemphigoides: four new cases and a review of the literature. Int J Dermatol. 2013;52:406-412. 
  2. Mohanarao TS, Kumar GA, Chennamsetty K, et al. Childhood lichen planus pemphigoides triggered by chickenpox. Indian Dermatol Online J. 2014;5:S98-S100. 
  3. Onprasert W, Chanprapaph K. Lichen planus pemphigoides induced by enalapril: a case report and a review of literature. Case Rep Dermatol. 2017;9:217-224. 
  4. Kuramoto N, Kishimoto S, Shibagaki R, et al. PUVA-induced lichen planus pemphigoides. Br J Dermatol. 2000;142:509-512. 
  5. Shimada H, Shono T, Sakai T, et al. Lichen planus pemphigoides concomitant with rectal adenocarcinoma: fortuitous or a true association? Eur J Dermatol. 2015;25:501-503. 
  6. Matos-Pires E, Campos S, Lencastre A, et al. Lichen planus pemphigoides. J Dtsch Dermatol Ges. 2018;16:335-337. 
  7. Zillikens D, Caux F, Mascaro JM, et al. Autoantibodies in lichen planus pemphigoides react with a novel epitope within the C-terminal NC16A domain of BP180. J Invest Dermatol. 1999;113:117-121. 
  8. Knisley RR, Petropolis AA, Mackey VT. Lichen planus pemphigoides treated with ustekinumab. Cutis. 2017;100:415-418. 
  9. Wagner G, Rose C, Sachse MM. Clinical variants of lichen planus. J Dtsch Dermatol Ges. 2013;11:309-319. 
  10. Bagci IS, Horvath ON, Ruzicka T, et al. Bullous pemphigoid. Autoimmun Rev. 2017;16:445-455. 
  11. Contestable JJ, Edhegard KD, Meyerle JH. Bullous systemic lupus erythematosus: a review and update to diagnosis and treatment. Am J Clin Dermatol. 2014;15:517-524. 
  12. Miller DD, Bhawan J. Bullous tinea pedis with direct immunofluorescence positivity: when is a positive result not autoimmune bullous disease? Am J Dermatopathol. 2013;35:587-594.
Issue
Cutis - 105(2)
Issue
Cutis - 105(2)
Page Number
62, 71-72
Page Number
62, 71-72
Publications
Publications
Topics
Article Type
Display Headline
Distinct Violaceous Plaques in Conjunction With Blisters
Display Headline
Distinct Violaceous Plaques in Conjunction With Blisters
Sections
Questionnaire Body

H&E, original magnification ×100

Direct immunofluorescence with IgG, original magnification ×400.

A 72-year-old woman presented to our dermatology clinic with a rash of several months' duration that began as itchy bumps on the wrists and spread to involve the legs. Approximately 2 months prior to presentation, she noted blisters on the feet and legs. She initially went to her primary care physician, who prescribed levofloxacin, cephalexin, and a 5-day course of prednisone. The prednisone initially helped; however the rash worsened on discontinuation. In our clinic, the patient had scattered tense bullae and numerous erosions with crust on the dorsum of the feet and legs, some of which were in conjunction with violaceous papules and plaques. There also was hypertrophic scale on the soles of the feet. A potassium hydroxide preparation of skin scrapings from the feet was negative for fungal elements. Two shave biopsies of a violaceous plaque and bulla as well as a perilesional punch biopsy from the leg were obtained.  

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Tue, 02/04/2020 - 11:45
Un-Gate On Date
Tue, 02/04/2020 - 11:45
Use ProPublica
CFC Schedule Remove Status
Tue, 02/04/2020 - 11:45
Hide sidebar & use full width
render the right sidebar.
Article PDF Media