Coalescing Hyperkeratotic Plaques and Papules

Article Type
Changed
Thu, 01/10/2019 - 13:55
Display Headline
Coalescing Hyperkeratotic Plaques and Papules

The Diagnosis: X-Linked Ichthyosis

Immunohistochemical staining of a punch biopsy specimen from the left foot with cytokeratin markers AE1/3, 5/6, and 19 showed normal positive uptake. Further workup was recommended, and the patient was referred to genetics for an ichthyosis gene panel. DNA sequencing revealed a c.1121G>A transition in exon 10 of the steroid sulfatase gene, STS, consistent with X-linked ichthyosis (XLI).

X-linked ichthyosis, also known as steroid sulfatase deficiency and X-linked recessive ichthyosis, is a congenital skin disorder classified in 1965 by Wells and Kerr.1 Ichthyoses are a heterogenous group of acquired and congenital disorders of keratinization that manifest with xerosis, hyperkeratosis, and scaling.2 Of more than 20 ichthyoses, XLI is the second most common ichthyosis, with a prevalence of 1 in 6000 males.3 X-linked ichthyosis occurs almost exclusively in males, and although females can be carriers, they rarely exhibit skin manifestations.4

X-linked ichthyosis is caused by either a partial or full deletion or mutation in the STS gene on the X chromosome.2 The absence of STS activity results in the accumulation of cholesterol sulfate in the stratum corneum, leading to corneocyte cohesion, hyperkeratosis, and impaired skin permeability. The most common clinical phenotype is characterized by polygonal scales concentrated on the upper and lower extremities as well as the trunk (Figure), consistent with our patient's clinical presentation.5

Figure
X-linked ichthyosis with platelike scaling and lichenified skin over the
anterior knees (A) as well as large exophytic papules on the upper
chest and neck (B).

X-linked ichthyosis typically presents in the first 6 months of life as generalized desquamation and xerosis that progresses to fine scaling on the trunk and extremities, more commonly and heavily involving the legs; however, the extensor surfaces of the arms also may be affected.6 After the neonatal period, fine scaling persists on the trunk and extremities, but scales often become coarser and darker over time. Although scaling is generalized, it typically spares the antecubital and popliteal fossae, palms, soles, and midface. The lateral face, axillae, and neck always remain involved.4 The most common extracutaneous manifestations of XLI affect the ocular, genitourinary, and cognitive/behavioral systems. Patients can develop corneal comma-shaped opacities, hypogonadism, cryptorchidism, and an increased risk for testicular cancer. Female carriers may have prolonged delivery of affected neonates.2,5,7-9 Given the unrelated debilitating neurologic consequences of our patient's presenting subarachnoid hemorrhage, further workup was not pursued into these associations.

Although XLI is most commonly diagnosed in early childhood, it also must be considered in adult patients presenting with severe scaling of the trunk, arms, and legs who have not had prior dermatologic workup. Given the similarity of XLI presentation to other ichthyoses, particularly ichthyosis vulgaris, lamellar ichthyosis, and ichthyosis bullosa of Siemens, genetic analysis is the most accurate diagnostic tool and should be considered in patients with an atypical presentation. Rupioid psoriasis also may be considered and can be confirmed on biopsy. Diagnosis of XLI should prompt symptomatic treatment, genetic counseling, and workup for extracutaneous manifestations.

References
  1. Wells RS, Kerr CB. Genetic classification of ichthyosis. Arch Dermatol. 1965;92:1-6.
  2. Fernandes NF, Janniger CK, Schwartz RA. X-linked ichthyosis: an oculocutaneous genodermatosis. J Am Acad Dermatol. 2010;62:480-485.
  3. Hernández-Martín A, González-Sarmiento R, De Unamuno P. X-linked ichthyosis: an update. Br J Dermatol. 1999;141:617-627.
  4. Elias PM, Williams ML, Choi EH, et al. Role of cholesterol sulfate in epidermal structure and function: lessons from X-linked ichthyosis [published online November 27, 2013]. Biochim Biophys Acta. 2014;1841:353-361.
  5. Wu B, Paller AS. Ichthyosis, X-Linked. Treasure Island, FL: StatPearls Publishing LLC; 2019.
  6. Marukian NV, Choate KA. Recent advances in understanding ichthyosis pathogenesis. F1000Res. 2016;5. doi:10.12688/f1000research.8584.1.
  7. Baek WS, Aypar U. Case report neurological manifestations of X-linked ichthyosis: case report and review of the literature [published online August 13, 2017]. 2017;2017:9086408.
  8. Brookes KJ, Hawi Z, Park J, et al. Polymorphisms of the steroid sulfatase (STS) gene are associated with attention deficit hyperactivity disorder and influence brain tissue mRNA expression. Am J Med Genet Part B Neuropsychiatr Genet. 2010;153:1417-1424.
  9. Kent L, Emerton J, Bhadravathi V, et al. X-linked ichthyosis (steroid sulfatase deficiency) is associated with increased risk of attention deficit hyperactivity disorder, autism and social communication deficits. J Med Genet. 2008;45:519-524.
Article PDF
Author and Disclosure Information

From the University of Illinois at Chicago. Ms. Jaros is from the College of Medicine, and Drs. Sergeyenko and Bain are from the Department of Dermatology.

The authors report no conflict of interest.

Correspondence: Joanna Jaros, BA, University of Illinois at Chicago, College of Medicine and Department of Dermatology, M/C 624, 808 S Wood St, 380 CME, Chicago, IL 60612-7307 (jjaros7@uic.edu).

Issue
Cutis - 102(6)
Publications
Topics
Page Number
402, 414-415
Sections
Author and Disclosure Information

From the University of Illinois at Chicago. Ms. Jaros is from the College of Medicine, and Drs. Sergeyenko and Bain are from the Department of Dermatology.

The authors report no conflict of interest.

Correspondence: Joanna Jaros, BA, University of Illinois at Chicago, College of Medicine and Department of Dermatology, M/C 624, 808 S Wood St, 380 CME, Chicago, IL 60612-7307 (jjaros7@uic.edu).

Author and Disclosure Information

From the University of Illinois at Chicago. Ms. Jaros is from the College of Medicine, and Drs. Sergeyenko and Bain are from the Department of Dermatology.

The authors report no conflict of interest.

Correspondence: Joanna Jaros, BA, University of Illinois at Chicago, College of Medicine and Department of Dermatology, M/C 624, 808 S Wood St, 380 CME, Chicago, IL 60612-7307 (jjaros7@uic.edu).

Article PDF
Article PDF
Related Articles

The Diagnosis: X-Linked Ichthyosis

Immunohistochemical staining of a punch biopsy specimen from the left foot with cytokeratin markers AE1/3, 5/6, and 19 showed normal positive uptake. Further workup was recommended, and the patient was referred to genetics for an ichthyosis gene panel. DNA sequencing revealed a c.1121G>A transition in exon 10 of the steroid sulfatase gene, STS, consistent with X-linked ichthyosis (XLI).

X-linked ichthyosis, also known as steroid sulfatase deficiency and X-linked recessive ichthyosis, is a congenital skin disorder classified in 1965 by Wells and Kerr.1 Ichthyoses are a heterogenous group of acquired and congenital disorders of keratinization that manifest with xerosis, hyperkeratosis, and scaling.2 Of more than 20 ichthyoses, XLI is the second most common ichthyosis, with a prevalence of 1 in 6000 males.3 X-linked ichthyosis occurs almost exclusively in males, and although females can be carriers, they rarely exhibit skin manifestations.4

X-linked ichthyosis is caused by either a partial or full deletion or mutation in the STS gene on the X chromosome.2 The absence of STS activity results in the accumulation of cholesterol sulfate in the stratum corneum, leading to corneocyte cohesion, hyperkeratosis, and impaired skin permeability. The most common clinical phenotype is characterized by polygonal scales concentrated on the upper and lower extremities as well as the trunk (Figure), consistent with our patient's clinical presentation.5

Figure
X-linked ichthyosis with platelike scaling and lichenified skin over the
anterior knees (A) as well as large exophytic papules on the upper
chest and neck (B).

X-linked ichthyosis typically presents in the first 6 months of life as generalized desquamation and xerosis that progresses to fine scaling on the trunk and extremities, more commonly and heavily involving the legs; however, the extensor surfaces of the arms also may be affected.6 After the neonatal period, fine scaling persists on the trunk and extremities, but scales often become coarser and darker over time. Although scaling is generalized, it typically spares the antecubital and popliteal fossae, palms, soles, and midface. The lateral face, axillae, and neck always remain involved.4 The most common extracutaneous manifestations of XLI affect the ocular, genitourinary, and cognitive/behavioral systems. Patients can develop corneal comma-shaped opacities, hypogonadism, cryptorchidism, and an increased risk for testicular cancer. Female carriers may have prolonged delivery of affected neonates.2,5,7-9 Given the unrelated debilitating neurologic consequences of our patient's presenting subarachnoid hemorrhage, further workup was not pursued into these associations.

Although XLI is most commonly diagnosed in early childhood, it also must be considered in adult patients presenting with severe scaling of the trunk, arms, and legs who have not had prior dermatologic workup. Given the similarity of XLI presentation to other ichthyoses, particularly ichthyosis vulgaris, lamellar ichthyosis, and ichthyosis bullosa of Siemens, genetic analysis is the most accurate diagnostic tool and should be considered in patients with an atypical presentation. Rupioid psoriasis also may be considered and can be confirmed on biopsy. Diagnosis of XLI should prompt symptomatic treatment, genetic counseling, and workup for extracutaneous manifestations.

The Diagnosis: X-Linked Ichthyosis

Immunohistochemical staining of a punch biopsy specimen from the left foot with cytokeratin markers AE1/3, 5/6, and 19 showed normal positive uptake. Further workup was recommended, and the patient was referred to genetics for an ichthyosis gene panel. DNA sequencing revealed a c.1121G>A transition in exon 10 of the steroid sulfatase gene, STS, consistent with X-linked ichthyosis (XLI).

X-linked ichthyosis, also known as steroid sulfatase deficiency and X-linked recessive ichthyosis, is a congenital skin disorder classified in 1965 by Wells and Kerr.1 Ichthyoses are a heterogenous group of acquired and congenital disorders of keratinization that manifest with xerosis, hyperkeratosis, and scaling.2 Of more than 20 ichthyoses, XLI is the second most common ichthyosis, with a prevalence of 1 in 6000 males.3 X-linked ichthyosis occurs almost exclusively in males, and although females can be carriers, they rarely exhibit skin manifestations.4

X-linked ichthyosis is caused by either a partial or full deletion or mutation in the STS gene on the X chromosome.2 The absence of STS activity results in the accumulation of cholesterol sulfate in the stratum corneum, leading to corneocyte cohesion, hyperkeratosis, and impaired skin permeability. The most common clinical phenotype is characterized by polygonal scales concentrated on the upper and lower extremities as well as the trunk (Figure), consistent with our patient's clinical presentation.5

Figure
X-linked ichthyosis with platelike scaling and lichenified skin over the
anterior knees (A) as well as large exophytic papules on the upper
chest and neck (B).

X-linked ichthyosis typically presents in the first 6 months of life as generalized desquamation and xerosis that progresses to fine scaling on the trunk and extremities, more commonly and heavily involving the legs; however, the extensor surfaces of the arms also may be affected.6 After the neonatal period, fine scaling persists on the trunk and extremities, but scales often become coarser and darker over time. Although scaling is generalized, it typically spares the antecubital and popliteal fossae, palms, soles, and midface. The lateral face, axillae, and neck always remain involved.4 The most common extracutaneous manifestations of XLI affect the ocular, genitourinary, and cognitive/behavioral systems. Patients can develop corneal comma-shaped opacities, hypogonadism, cryptorchidism, and an increased risk for testicular cancer. Female carriers may have prolonged delivery of affected neonates.2,5,7-9 Given the unrelated debilitating neurologic consequences of our patient's presenting subarachnoid hemorrhage, further workup was not pursued into these associations.

Although XLI is most commonly diagnosed in early childhood, it also must be considered in adult patients presenting with severe scaling of the trunk, arms, and legs who have not had prior dermatologic workup. Given the similarity of XLI presentation to other ichthyoses, particularly ichthyosis vulgaris, lamellar ichthyosis, and ichthyosis bullosa of Siemens, genetic analysis is the most accurate diagnostic tool and should be considered in patients with an atypical presentation. Rupioid psoriasis also may be considered and can be confirmed on biopsy. Diagnosis of XLI should prompt symptomatic treatment, genetic counseling, and workup for extracutaneous manifestations.

References
  1. Wells RS, Kerr CB. Genetic classification of ichthyosis. Arch Dermatol. 1965;92:1-6.
  2. Fernandes NF, Janniger CK, Schwartz RA. X-linked ichthyosis: an oculocutaneous genodermatosis. J Am Acad Dermatol. 2010;62:480-485.
  3. Hernández-Martín A, González-Sarmiento R, De Unamuno P. X-linked ichthyosis: an update. Br J Dermatol. 1999;141:617-627.
  4. Elias PM, Williams ML, Choi EH, et al. Role of cholesterol sulfate in epidermal structure and function: lessons from X-linked ichthyosis [published online November 27, 2013]. Biochim Biophys Acta. 2014;1841:353-361.
  5. Wu B, Paller AS. Ichthyosis, X-Linked. Treasure Island, FL: StatPearls Publishing LLC; 2019.
  6. Marukian NV, Choate KA. Recent advances in understanding ichthyosis pathogenesis. F1000Res. 2016;5. doi:10.12688/f1000research.8584.1.
  7. Baek WS, Aypar U. Case report neurological manifestations of X-linked ichthyosis: case report and review of the literature [published online August 13, 2017]. 2017;2017:9086408.
  8. Brookes KJ, Hawi Z, Park J, et al. Polymorphisms of the steroid sulfatase (STS) gene are associated with attention deficit hyperactivity disorder and influence brain tissue mRNA expression. Am J Med Genet Part B Neuropsychiatr Genet. 2010;153:1417-1424.
  9. Kent L, Emerton J, Bhadravathi V, et al. X-linked ichthyosis (steroid sulfatase deficiency) is associated with increased risk of attention deficit hyperactivity disorder, autism and social communication deficits. J Med Genet. 2008;45:519-524.
References
  1. Wells RS, Kerr CB. Genetic classification of ichthyosis. Arch Dermatol. 1965;92:1-6.
  2. Fernandes NF, Janniger CK, Schwartz RA. X-linked ichthyosis: an oculocutaneous genodermatosis. J Am Acad Dermatol. 2010;62:480-485.
  3. Hernández-Martín A, González-Sarmiento R, De Unamuno P. X-linked ichthyosis: an update. Br J Dermatol. 1999;141:617-627.
  4. Elias PM, Williams ML, Choi EH, et al. Role of cholesterol sulfate in epidermal structure and function: lessons from X-linked ichthyosis [published online November 27, 2013]. Biochim Biophys Acta. 2014;1841:353-361.
  5. Wu B, Paller AS. Ichthyosis, X-Linked. Treasure Island, FL: StatPearls Publishing LLC; 2019.
  6. Marukian NV, Choate KA. Recent advances in understanding ichthyosis pathogenesis. F1000Res. 2016;5. doi:10.12688/f1000research.8584.1.
  7. Baek WS, Aypar U. Case report neurological manifestations of X-linked ichthyosis: case report and review of the literature [published online August 13, 2017]. 2017;2017:9086408.
  8. Brookes KJ, Hawi Z, Park J, et al. Polymorphisms of the steroid sulfatase (STS) gene are associated with attention deficit hyperactivity disorder and influence brain tissue mRNA expression. Am J Med Genet Part B Neuropsychiatr Genet. 2010;153:1417-1424.
  9. Kent L, Emerton J, Bhadravathi V, et al. X-linked ichthyosis (steroid sulfatase deficiency) is associated with increased risk of attention deficit hyperactivity disorder, autism and social communication deficits. J Med Genet. 2008;45:519-524.
Issue
Cutis - 102(6)
Issue
Cutis - 102(6)
Page Number
402, 414-415
Page Number
402, 414-415
Publications
Publications
Topics
Article Type
Display Headline
Coalescing Hyperkeratotic Plaques and Papules
Display Headline
Coalescing Hyperkeratotic Plaques and Papules
Sections
Questionnaire Body

A 67-year-old man with a history of congestive heart failure, type 2 diabetes mellitus, hypertension, and schizophrenia was admitted to the hospital for subarachnoid hemorrhage and was noted to have heavy scaling on the bilateral legs. Given recent medical events, the patient was nonconversant at the time of consultation, but his daughter provided his medical history at bedside. The patient usually wore long-sleeved clothing and pants, thus no one had seen his skin in many years, and it was unclear how long the scaling had been present. His family history was notable for eczema in distant relatives but negative for comparable conditions. Physical examination revealed thick lichenified skin with many large, exophytic, brown papules (largest measured 1.5×1×1 cm) and platelike scaling on the anterior chest, abdomen, lateral arms, and forearms. Extensive coalescing hyperkeratotic plaques and papules (up to 1 cm in thickness) were present on the anterior legs and feet, and scattered verrucous brown papules were noted on the plantar aspects of the bilateral feet. A punch biopsy of the left foot revealed extensive, dense, compact, orthokeratotic hyperkeratosis with a preserved granular layer with no epidermolysis.

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Mon, 11/26/2018 - 10:00
Un-Gate On Date
Mon, 11/26/2018 - 10:00
Use ProPublica
CFC Schedule Remove Status
Mon, 11/26/2018 - 10:00
Article PDF Media

Montreal Cognitive Assessment fares well for rapid, reliable screening in SLE

Article Type
Changed
Sat, 12/08/2018 - 15:30

 

– The Montreal Cognitive Assessment Test provides persuasive advantages over the standard neuropsychological test battery often recommended in guidelines as a screening tool for cognitive impairment in patients with systemic lupus erythematosus, Nicolas Paez-Venegas, MD, asserted at the annual meeting of the American College of Rheumatology.

The MoCA, as it’s known, offers brevity, simplicity, and none of the considerable expense and inconvenience of bringing in a trained specialist to administer a neuropsychological battery. Moreover, in a comparative efficacy study, the MoCA outperformed two other brief screening tools for cognitive impairment – the Mini-Mental State Examination and the Cognitive Symptom Inventory – and showed excellent correspondence with the results of the formal neuropsychological battery, reported Dr. Paez-Venegas, a psychiatrist at the Jalisco Institute of Mental Health, in Zapopan, Mexico.

He presented a cross-sectional study that pitted the three brief screening tests against a gold-standard neuropsychological battery in 44 patients with systemic lupus erythematosus (SLE) according to the 2012 Systemic Lupus International Collaborating Clinics Criteria, none of whom had any known medical or psychiatric comorbidities.

The MoCA proved to have the best congruence with the findings of the neuropsychological battery, with an area under the curve of 99.4%, 84% sensitivity, and 100% specificity for cognitive impairment. The Mini-Mental State Examination had 55% sensitivity and 100% specificity, while the Cognitive Symptom Inventory displayed 55% sensitivity and 31% specificity.

“We therefore encourage rheumatologists to apply the MoCA test as a valuable and easily implemented tool for detecting cognitive impairment as part of an integrated approach in SLE,” Dr. Paez-Venegas said.

Periodic screening for cognitive impairment in patients with SLE is an important aspect of patient management because cognitive impairment is a common manifestation of the disease, affecting up to two-thirds of patients, and it can have a serious impact upon quality of life and self-concept. Because such screening isn’t a one-time event, resort to a neuropsychological battery becomes particularly problematic. The battery employed in this study included the Wechsler Adult Intelligence Scale–Fourth Edition test, the Digit-Symbol test, the Finger-Tapping test of motor control, the Stroop test, Trail Making A and B, the Paced Auditory Serial Addition test, letter-number sequencing, the Wechsler Vocabulary test, the Rey-Osterrieth complex figure test, semantic and phonemic fluency tests, and a test of verbal Spanish comprehension. The battery is a comprehensive tool often employed in research studies but is not well suited for use in a busy clinical practice.

Overall, 70% of the SLE patients demonstrated cognitive impairment in one or more domains on the neuropsychological battery. Processing speed was the most frequently affected domain, involving 23 of the 44 patients. Only a single patient displayed abnormal motor control.

The MoCA test assesses attention, executive function, concentration, language, memory, abstraction, orientation, visuospatial cognitive capacity, and calculation.

Dr. Paez-Venegas’s study was published online earlier this year (J Clin Rheumatol 2018 Jul 18. doi: 10.1097/RHU.0000000000000876). He reported having no financial conflicts regarding his study.

SOURCE: Paez-Venegas N et al. Arthritis Rheumatol. 2018;70(Suppl 10): Abstract 708.

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

 

– The Montreal Cognitive Assessment Test provides persuasive advantages over the standard neuropsychological test battery often recommended in guidelines as a screening tool for cognitive impairment in patients with systemic lupus erythematosus, Nicolas Paez-Venegas, MD, asserted at the annual meeting of the American College of Rheumatology.

The MoCA, as it’s known, offers brevity, simplicity, and none of the considerable expense and inconvenience of bringing in a trained specialist to administer a neuropsychological battery. Moreover, in a comparative efficacy study, the MoCA outperformed two other brief screening tools for cognitive impairment – the Mini-Mental State Examination and the Cognitive Symptom Inventory – and showed excellent correspondence with the results of the formal neuropsychological battery, reported Dr. Paez-Venegas, a psychiatrist at the Jalisco Institute of Mental Health, in Zapopan, Mexico.

He presented a cross-sectional study that pitted the three brief screening tests against a gold-standard neuropsychological battery in 44 patients with systemic lupus erythematosus (SLE) according to the 2012 Systemic Lupus International Collaborating Clinics Criteria, none of whom had any known medical or psychiatric comorbidities.

The MoCA proved to have the best congruence with the findings of the neuropsychological battery, with an area under the curve of 99.4%, 84% sensitivity, and 100% specificity for cognitive impairment. The Mini-Mental State Examination had 55% sensitivity and 100% specificity, while the Cognitive Symptom Inventory displayed 55% sensitivity and 31% specificity.

“We therefore encourage rheumatologists to apply the MoCA test as a valuable and easily implemented tool for detecting cognitive impairment as part of an integrated approach in SLE,” Dr. Paez-Venegas said.

Periodic screening for cognitive impairment in patients with SLE is an important aspect of patient management because cognitive impairment is a common manifestation of the disease, affecting up to two-thirds of patients, and it can have a serious impact upon quality of life and self-concept. Because such screening isn’t a one-time event, resort to a neuropsychological battery becomes particularly problematic. The battery employed in this study included the Wechsler Adult Intelligence Scale–Fourth Edition test, the Digit-Symbol test, the Finger-Tapping test of motor control, the Stroop test, Trail Making A and B, the Paced Auditory Serial Addition test, letter-number sequencing, the Wechsler Vocabulary test, the Rey-Osterrieth complex figure test, semantic and phonemic fluency tests, and a test of verbal Spanish comprehension. The battery is a comprehensive tool often employed in research studies but is not well suited for use in a busy clinical practice.

Overall, 70% of the SLE patients demonstrated cognitive impairment in one or more domains on the neuropsychological battery. Processing speed was the most frequently affected domain, involving 23 of the 44 patients. Only a single patient displayed abnormal motor control.

The MoCA test assesses attention, executive function, concentration, language, memory, abstraction, orientation, visuospatial cognitive capacity, and calculation.

Dr. Paez-Venegas’s study was published online earlier this year (J Clin Rheumatol 2018 Jul 18. doi: 10.1097/RHU.0000000000000876). He reported having no financial conflicts regarding his study.

SOURCE: Paez-Venegas N et al. Arthritis Rheumatol. 2018;70(Suppl 10): Abstract 708.

 

– The Montreal Cognitive Assessment Test provides persuasive advantages over the standard neuropsychological test battery often recommended in guidelines as a screening tool for cognitive impairment in patients with systemic lupus erythematosus, Nicolas Paez-Venegas, MD, asserted at the annual meeting of the American College of Rheumatology.

The MoCA, as it’s known, offers brevity, simplicity, and none of the considerable expense and inconvenience of bringing in a trained specialist to administer a neuropsychological battery. Moreover, in a comparative efficacy study, the MoCA outperformed two other brief screening tools for cognitive impairment – the Mini-Mental State Examination and the Cognitive Symptom Inventory – and showed excellent correspondence with the results of the formal neuropsychological battery, reported Dr. Paez-Venegas, a psychiatrist at the Jalisco Institute of Mental Health, in Zapopan, Mexico.

He presented a cross-sectional study that pitted the three brief screening tests against a gold-standard neuropsychological battery in 44 patients with systemic lupus erythematosus (SLE) according to the 2012 Systemic Lupus International Collaborating Clinics Criteria, none of whom had any known medical or psychiatric comorbidities.

The MoCA proved to have the best congruence with the findings of the neuropsychological battery, with an area under the curve of 99.4%, 84% sensitivity, and 100% specificity for cognitive impairment. The Mini-Mental State Examination had 55% sensitivity and 100% specificity, while the Cognitive Symptom Inventory displayed 55% sensitivity and 31% specificity.

“We therefore encourage rheumatologists to apply the MoCA test as a valuable and easily implemented tool for detecting cognitive impairment as part of an integrated approach in SLE,” Dr. Paez-Venegas said.

Periodic screening for cognitive impairment in patients with SLE is an important aspect of patient management because cognitive impairment is a common manifestation of the disease, affecting up to two-thirds of patients, and it can have a serious impact upon quality of life and self-concept. Because such screening isn’t a one-time event, resort to a neuropsychological battery becomes particularly problematic. The battery employed in this study included the Wechsler Adult Intelligence Scale–Fourth Edition test, the Digit-Symbol test, the Finger-Tapping test of motor control, the Stroop test, Trail Making A and B, the Paced Auditory Serial Addition test, letter-number sequencing, the Wechsler Vocabulary test, the Rey-Osterrieth complex figure test, semantic and phonemic fluency tests, and a test of verbal Spanish comprehension. The battery is a comprehensive tool often employed in research studies but is not well suited for use in a busy clinical practice.

Overall, 70% of the SLE patients demonstrated cognitive impairment in one or more domains on the neuropsychological battery. Processing speed was the most frequently affected domain, involving 23 of the 44 patients. Only a single patient displayed abnormal motor control.

The MoCA test assesses attention, executive function, concentration, language, memory, abstraction, orientation, visuospatial cognitive capacity, and calculation.

Dr. Paez-Venegas’s study was published online earlier this year (J Clin Rheumatol 2018 Jul 18. doi: 10.1097/RHU.0000000000000876). He reported having no financial conflicts regarding his study.

SOURCE: Paez-Venegas N et al. Arthritis Rheumatol. 2018;70(Suppl 10): Abstract 708.

Publications
Publications
Topics
Article Type
Sections
Article Source

REPORTING FROM THE ACR ANNUAL MEETING

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

 

Key clinical point: The Montreal Cognitive Assessment Test is a useful tool for screening for cognitive impairment in systemic lupus erythematosus.

Major finding: The Montreal Cognitive Assessment Test showed 84% sensitivity and 100% specificity for cognitive impairment in systemic lupus erythematosus patients.

Study details: This comparative effectiveness study included 44 systemic lupus erythematosus patients assessed for cognitive impairment using three different tools.

Disclosures: The presenter reported having no financial conflicts regarding this study, which was conducted free of commercial support.

Source: Paez-Venegas N et al. Arthritis Rheumatol. 2018;70(Suppl 10): Abstract 708.

Disqus Comments
Default
Use ProPublica

Update on Calciphylaxis Etiopathogenesis, Diagnosis, and Management

Article Type
Changed
Fri, 10/25/2019 - 11:15
Display Headline
Update on Calciphylaxis Etiopathogenesis, Diagnosis, and Management
In partnership with the Society for Dermatology Hospitalists

Calciphylaxis, also known as calcific uremic arteriolopathy, is a painful skin condition classically seen in patients with end-stage renal disease (ESRD), particularly those on chronic dialysis.1,2 It also has increasingly been reported in patients with normal renal function and calcium and phosphate homeostasis.3,4 Effective diagnosis and management of calciphylaxis remains challenging for physicians.2,5 The condition is characterized by tissue ischemia caused by calcification of cutaneous arteriolar vessels. As a result, calciphylaxis is associated with high mortality rates, ranging from 60% to 80%.5,6 Excruciating pain and nonhealing ulcers often lead to recurrent hospitalizations and infectious complications,7 and poor nutritional status, chronic pain, depression, and insomnia can further complicate recovery and lead to poor quality of life.8

We provide an update on calciphylaxis etiopathogenesis, diagnosis, and management. We also highlight some challenges faced in managing this potentially fatal condition.

Epidemiology

Calciphylaxis is considered a rare dermatosis with an estimated annual incidence of 1% to 4% in ESRD patients on dialysis. Recent data suggest that incidence of calciphylaxis is rising,5,7,9 which may stem from an increased use of calcium-based phosphate binders, an actual rise in disease incidence, and/or increased recognition of the disease.5 It is difficult to estimate the exact disease burden of calciphylaxis because the diagnostic criteria are not well defined, often leading to missed or delayed diagnosis.3,10 Furthermore, there is no centralized registry for calciphylaxis cases.3

Etiology and Pathogenesis

Calciphylaxis is thought to have a multifactorial etiology with the exact cause or trigger unknown.7 A long list of risk factors and triggers is associated with the condition (Table 1). Calciphylaxis primarily affects small arteries (40–600 μm in diameter) that become calcified due to an imbalance between inhibitors and promoters of calcification.2,11 Fetuin-A and matrix Gla protein inhibit vascular calcification and are downregulated in calciphylaxis.12,13 Dysfunctional calcium, phosphate, and parathyroid hormone regulatory pathways provide an increased substrate for the process of calcification, which causes endothelial damage and microthrombosis, resulting in tissue ischemia and infarction.14,15 Notably, there is growing interest in the role of vitamin K in the pathogenesis of calciphylaxis. Vitamin K inhibits vascular calcification, possibly by increasing the circulating levels of carboxylated matrix Gla protein.16

Clinical Features

Calciphylaxis is most commonly seen on the legs, abdomen, and buttocks.2 Patients with ESRD commonly develop proximal lesions affecting adipose-rich sites and have a poor prognosis. Distal lesions are more common in patients with nonuremic calciphylaxis, and mortality rates are lower in this population.2

Early lesions present as painful skin nodules or indurated plaques that often are rock-hard or firm to palpation with overlying mottling or a livedoid pattern (Figure, A). Early lesions progress from livedo reticularis to livedo racemosa and then to retiform purpura (Figure, B). Purpuric lesions later evolve into black eschars (Figure, C), then to necrotic, ulcerated, malodorous plaques or nodules in later stages of the disease (Figure, D). Lesions also may develop a gangrenous sclerotic appearance.2,5

Figure
Early lesions of calciphylaxis often appear as indurated plaques with overlying mottling or livedoid pattern (A) that progress to retiform purpura (B). Purpuric lesions then evolve into black eschars (C). In later stages, necrotic, ulcerated, malodorous plaques or nodules are present (D).

Although most patients with calciphylaxis have ESRD, nonuremic patients also can develop the disease. Those with calciphylaxis who do not have renal dysfunction frequently have other risk factors for the disease and often report another notable health problem in the weeks or months prior to presentation.4 More than half of patients with calciphylaxis become bedridden or require use of a wheelchair.17 Pain is characteristically severe throughout the course of the disease; it may even precede the appearance of the skin lesions.18 Because the pain is associated with ischemia, it tends to be relatively refractory to treatment with opioids. Rare extracutaneous vascular calcifications may lead to visual impairment, gastrointestinal tract bleeding, and myopathy.5,9,19,20

Diagnosis

Considering the high morbidity and mortality associated with calciphylaxis, it is important to provide accurate and timely diagnosis; however, there currently are no validated diagnostic criteria for calciphylaxis. Careful correlation of clinical and histologic findings is required. Calciphylaxis biopsies have demonstrated medial calcification and proliferation of the intima of small- to medium-sized arteries.21 Lobular and septal panniculitis and extravascular soft-tissue calcification, particularly stippled calcification of the eccrine sweat glands, also has been seen.2,22 Special calcium stains (eg, von Kossa, Alizarin red) increase the sensitivity of biopsy by highlighting subtle areas of intravascular and extravascular calcification.5,23 Sufficient sampling of subcutaneous tissue and specimen evaluation by an experienced dermatopathologist are necessary to ensure proper interpretation of the histologic findings.

Despite these measures, skin biopsies may be nondiagnostic or falsely negative; therefore, when there is high clinical suspicion, it may be appropriate to move forward with a presumptive diagnosis of calciphylaxis even if the histologic findings are nondiagnostic.1,9,24 It also is worth noting that localized progression and ulceration may occur following skin biopsy, such that biopsy may even be contraindicated in certain cases (eg, penile calciphylaxis).

Standard laboratory workup for calciphylaxis includes evaluation for associated risk factors as well as exclusion of other conditions in the differential diagnosis (Table 2). Blood tests to evaluate for risk factors include liver and renal function tests, a complete metabolic panel, parathyroid hormone level, and serum albumin level.5 Elevated calcium and phosphate levels may signal disturbed calcium and phosphate homeostasis but are neither sensitive nor specific for the diagnosis.25 Complete blood cell count, blood cultures, thorough hypercoagulability workup (including but not limited to antiphospholipid antibodies, proteins C and S, factor V Leiden, antithrombin III, homocysteine, methylenetetrahydrofolate reductase mutation, and cryoglobulins), rheumatoid factor, antineutrophil cytoplasmic antibodies, and antinuclear antibody testing may be relevant to help identify contributing factors or mimickers of calciphylaxis.5 Various imaging modalities also have been used to evaluate for the presence of soft-tissue calcification in areas of suspected calciphylaxis, including radiography, mammography, computed tomography, ultrasonography, nuclear bone scintigraphy, and spectroscopy.2,26,27 Unfortunately, there currently is no standardized reproducible imaging modality for reliable diagnosis of calciphylaxis. Ultimately, histologic and radiographic findings should always be interpreted in the context of relevant clinical findings.2,9

 

 

Prevention

Reduction of the net calcium phosphorus product may help reduce the risk of calciphylaxis in ESRD patients, which can be accomplished by using non–calcium-phosphate binders, adequate dialysis, and restricting use of vitamin D and vitamin K antagonists.2,5 There are limited data regarding the benefits of using bisphosphonates and cinacalcet in ESRD patients on dialysis to prevent calciphylaxis.28,29

Management

Management of calciphylaxis is multifactorial. Besides dermatology and nephrology, specialists in pain management, wound care, plastic surgery, and nutrition are critical partners in management.1,5,9,30 Nephrologists can help optimize calcium and phosphate balance and ensure adequate dialysis. Pain specialists can aid in creating aggressive multiagent pain regimens that target the neuropathic/ischemic and physical aspects of calciphylaxis pain. When appropriate, nutrition specialists can help establish high-protein, low-phosphorus diets, and wound specialists can provide access to advanced wound dressings and adjunctive hyperbaric oxygen therapy. Plastic surgeons can provide conservative debridement procedures in a subset of patients, usually those with distal stable disease.

The limited understanding of the etiopathogenesis of calciphylaxis and the lack of data on its management are reflected in the limited treatment options for the disease (Table 3).2,5,9 There are no formal algorithms for the treatment of calciphylaxis. Therapeutic trials are scarce, and most of the current treatment recommendations are based on small retrospective reports or case series. Sodium thiosulfate has been the most widely used treatment option since 2004, when its use in calciphylaxis was first reported.31 Sodium thiosulfate chelates calcium and is thought to have antioxidant and vasodilatory properties.32 There are a few promising clinical trials and large-scale studies (Table 4) that aim to evaluate the efficacy of existing treatments (eg, sodium thiosulfate) as well as novel treatment options such as lanthanum carbonate, SNF472 (hexasodium phytate), and vitamin K.33-36

Prognosis

Calciphylaxis is a potentially fatal condition with a poor prognosis and a median survival rate of approximately 1 year following the appearance of skin lesions.37-39 Patients with proximal lesions and those on peritoneal dialysis (as opposed to hemodialysis) have a worse prognosis.40 Mortality rates are estimated to be 30% at 6 months, 50% at 12 months, and 80% at 2 years, with sepsis secondary to infection of cutaneous ulcers being the leading cause of death.37-39 The impact of calciphylaxis on patient quality of life and activities of daily living is severe.8,17

Future Directions

Multi-institution cohort studies and collaborative registries are needed to provide updated information related to the epidemiology, diagnosis, treatment, morbidity, and mortality associated with calciphylaxis and to help formulate evidence-based diagnostic criteria. Radiographic and histologic studies, as well as other tools for early and accurate diagnosis of calciphylaxis, should be studied for feasibility, accuracy, and reproducibility. The incidence of nonuremic calciphylaxis points toward pathogenic pathways besides those based on the bone-mineral axis. Basic science research directed at improving understanding of the pathophysiology of calciphylaxis would be helpful in devising new treatment strategies targeting these pathways. Establishment of a collaborative, multi-institutional calciphylaxis working group would enable experts to formulate therapeutic guidelines based on current evidence. Such a group could facilitate initiation of large prospective studies to establish the efficacy of existing and new treatment modalities for calciphylaxis. A working group within the Society for Dermatology Hospitalists has been tasked with addressing these issues and is currently establishing a multicenter calciphylaxis database.

References
  1. Nigwekar SU, Kroshinsky D, Nazarian RM, et al. Calciphylaxis: risk factors, diagnosis, and treatment. Am J Kidney Dis. 2015;66:133-146.
  2. Nigwekar SU, Thadhani RI, Brandenburg VM. Calciphylaxis. N Engl J Med. 2018;378:1704-1714.
  3. Davis JM. The relationship between obesity and calciphylaxis: a review of the literature. Ostomy Wound Manage. 2016;62:12-18.
  4. Bajaj R, Courbebaisse M, Kroshinsky D, et al. Calciphylaxis in patients with normal renal function: a case series and systematic review. Mayo Clin Proc. 2018;93:1202-1212.
  5. Hafner J, Keusch G, Wahl C, et al. Uremic small-artery disease with medial calcification and intimal hyperplasia (so-called calciphylaxis): a complication of chronic renal failure and benefit from parathyroidectomy. J Am Acad Dermatol. 1995;33:954-962.
  6. Jeong HS, Dominguez AR. Calciphylaxis: controversies in pathogenesis, diagnosis and treatment. Am J Med Sci. 2016;351:217-227.
  7. Westphal SG, Plumb T. Calciphylaxis. In: StatPearls. Treasure Island, FL: StatPearls Publishing; 2018. https://www.ncbi.nlm.nih.gov/books/NBK519020. Accessed November 12, 2018.
  8. Riemer CA, El-Azhary RA, Wu KL, et al. Underreported use of palliative care and patient-reported outcome measures to address reduced quality of life in patients with calciphylaxis: a systematic review. Br J Dermatol. 2017;177:1510-1518.
  9. Nigwekar SU. Calciphylaxis. Curr Opin Nephrol Hypertens. 2017;26:276-281.
  10. Fine A, Fontaine B. Calciphylaxis: the beginning of the end? Perit Dial Int. 2008;28:268-270.
  11. Lin WT, Chao CM. Tumoral calcinosis in renal failure. QJM. 2014;107:387.
  12. Schafer C, Heiss A, Schwarz A, et al. The serum protein alpha 2-Heremans-Schmid glycoprotein/fetuin-A is a systemically acting inhibitor of ectopic calcification. J Clin Invest. 2003;112:357-366.
  13. Luo G, Ducy P, McKee MD, et al. Spontaneous calcification of arteries and cartilage in mice lacking matrix GLA protein. Nature. 1997;386:78-81.
  14. Bleyer AJ, Choi M, Igwemezie B, et al. A case control study of proximal calciphylaxis. Am J Kidney Dis. 1998;32:376-383.
  15. Ahmed S, O’Neill KD, Hood AF, et al. Calciphylaxis is associated with hyperphosphatemia and increased osteopontin expression by vascular smooth muscle cells. Am J Kidney Dis. 2001;37:267-276.
  16. Nigwekar SU, Bloch DB, Nazarian RM, et al. Vitamin K-dependent carboxylation of matrix gla protein influences the risk of calciphylaxis. J Am Soc Nephrol. 2017;28:1717-1722.
  17. Weenig RH, Sewell LD, Davis MD, et al. Calciphylaxis: natural history, risk factor analysis, and outcome. J Am Acad Dermatol. 2007;56:569-579.
  18. Polizzotto MN, Bryan T, Ashby MA, et al. Symptomatic management of calciphylaxis: a case series and review of the literature. J Pain Symptom Manage. 2006;32:186-190.
  19. Gupta N, Haq KF, Mahajan S, et al. Gastrointestinal bleeding secondary to calciphylaxis. Am J Case Rep. 2015;16:818-822.
  20. Edelstein CL, Wickham MK, Kirby PA. Systemic calciphylaxis presenting as a painful, proximal myopathy. Postgrad Med J. 1992;68:209-211.
  21. Mochel MC, Arakari RY, Wang G, et al. Cutaneous calciphylaxis: a retrospective histopathologic evaluation. Am J Dermatopathol. 2013;35:582-586.
  22. Chen TY, Lehman JS, Gibson LE, et al. Histopathology of calciphylaxis: cohort study with clinical correlations. Am J Dermatopathol. 2017;39:795-802.
  23. Cassius C, Moguelet P, Monfort JB, et al. Calciphylaxis in haemodialysed patients: diagnostic value of calcifications in cutaneous biopsy. Br J Dermatol. 2018;178:292-293.
  24. Sreedhar A, Sheikh HA, Scagliotti CJ, et al. Advanced-stage calciphylaxis: think before you punch. Cleve Clin J Med. 2016;83:562-564.
  25. Brandenburg VM, Kramann R, Rothe H, et al. Calcific uraemic arteriolopathy (calciphylaxis): data from a large nation-wide registry. Nephrol Dial Transplant. 2017;32:126-132.
  26. Paul S, Rabito CA, Vedak P, et al. The role of bone scintigraphy in the diagnosis of calciphylaxis. JAMA Dermatol. 2017;153:101-103.
  27. Shmidt E, Murthy NS, Knudsen JM, et al. Net-like pattern of calcification on plain soft-tissue radiographs in patients with calciphylaxis. J Am Acad Dermatol. 2012;67:1296-1301.
  28. EVOLVE Trial Investigators; Chertow GM, Block GA, Correa-Rotter R, et al. Effect of cinacalcet on cardiovascular disease in patients undergoing dialysis. N Engl J Med. 2012;367:2482-2494.
  29. Rogers NM, Teubner DJO, Coates PT. Calcific uremic arteriolopathy: advances in pathogenesis and treatment. Semin Dial. 2007;20:150-157.
  30. Nigwekar SU. Multidisciplinary approach to calcific uremic arteriolopathy. Curr Opin Nephrol Hypertens. 2015;24:531-537.
  31. Cicone JS, Petronis JB, Embert CD, et al. Successful treatment of calciphylaxis with intravenous sodium thiosulfate. Am J Kidney Dis. 2004;43:1104-1108.
  32. Chen NX, O’Neill K, Akl NK, et al. Adipocyte induced arterial calcification is prevented with sodium thiosulfate. Biochem Biophys Res Commun. 2014;449:151-156.
  33. Chan MR, Ghandour F, Murali NS, et al. Pilot study of the effect of lanthanum carbonate in patients with calciphylaxis: a Wisconsin Network for Health Research (WiNHR) study. J Nephrol Ther. 2014;4:1000162.
  34. Perelló J, Gómez M, Ferrer MD, et al. SNF472, a novel inhibitor of vascular calcification, could be administered during hemodialysis to attain potentially therapeutic phytate levels. J Nephrol. 2018;31:287-296.
  35. Christiadi D, Singer RF. Calciphylaxis in a dialysis patient successfully treated with high-dose vitamin K supplementation. Clin Kidney J. 2018;11:528-529.
  36. Caluwe R, Vandecasteele S, Van Vlem B, et al. Vitamin K2 supplementation in haemodialysis patients: a randomized dose-finding study. Nephrol Dial Transplant. 2014;29:1385-1390.
  37. McCarthy JT, El-Azhary RA, Patzelt MT, et al. Survival, risk factors, and effect of treatment in 101 patients with calciphylaxis. Mayo Clin Proc. 2016;91:1384-1394.
  38. Fine A, Zacharias J. Calciphylaxis is usually non-ulcerating: risk factors, outcome and therapy. Kidney Int. 2002;61:2210-2217.
  39. Nigwekar SU, Zhao S, Wenger J, et al. A nationally representative study of calcific uremic arteriolopathy risk factors. J Am Soc Nephrol. 2016;27:3421-3429.
  40. Zhang Y, Corapi KM, Luongo M, et al. Calciphylaxis in peritoneal dialysis patients: a single center cohort study. Int J Nephrol Renovasc Dis. 2016;9:235-241.
Article PDF
Author and Disclosure Information

Dr. Khanna is from the Department of Dermatology, Cleveland Clinic, Ohio. Dr. Dominguez is from the Department of Dermatology, University of Texas Southwestern Medical Center, Dallas. Drs. Keller and Ortega-Loayza are from the Department of Dermatology, Oregon Health & Science University, Portland. Dr. Kroshinsky is from the Department of Dermatology, Massachusetts General Hospital, Boston. Dr. Strowd is from the Department of Dermatology, Wake Forest University School of Medicine, Winston-Salem, North Carolina. Dr. Micheletti is from the Departments of Dermatology and Medicine, University of Pennsylvania, Philadelphia.

The authors report no conflict of interest.

Correspondence: Robert G. Micheletti, MD (Robert.Micheletti@uphs.upenn.edu).

Issue
Cutis - 102(6)
Publications
Topics
Page Number
395-400
Sections
Author and Disclosure Information

Dr. Khanna is from the Department of Dermatology, Cleveland Clinic, Ohio. Dr. Dominguez is from the Department of Dermatology, University of Texas Southwestern Medical Center, Dallas. Drs. Keller and Ortega-Loayza are from the Department of Dermatology, Oregon Health & Science University, Portland. Dr. Kroshinsky is from the Department of Dermatology, Massachusetts General Hospital, Boston. Dr. Strowd is from the Department of Dermatology, Wake Forest University School of Medicine, Winston-Salem, North Carolina. Dr. Micheletti is from the Departments of Dermatology and Medicine, University of Pennsylvania, Philadelphia.

The authors report no conflict of interest.

Correspondence: Robert G. Micheletti, MD (Robert.Micheletti@uphs.upenn.edu).

Author and Disclosure Information

Dr. Khanna is from the Department of Dermatology, Cleveland Clinic, Ohio. Dr. Dominguez is from the Department of Dermatology, University of Texas Southwestern Medical Center, Dallas. Drs. Keller and Ortega-Loayza are from the Department of Dermatology, Oregon Health & Science University, Portland. Dr. Kroshinsky is from the Department of Dermatology, Massachusetts General Hospital, Boston. Dr. Strowd is from the Department of Dermatology, Wake Forest University School of Medicine, Winston-Salem, North Carolina. Dr. Micheletti is from the Departments of Dermatology and Medicine, University of Pennsylvania, Philadelphia.

The authors report no conflict of interest.

Correspondence: Robert G. Micheletti, MD (Robert.Micheletti@uphs.upenn.edu).

Article PDF
Article PDF
In partnership with the Society for Dermatology Hospitalists
In partnership with the Society for Dermatology Hospitalists

Calciphylaxis, also known as calcific uremic arteriolopathy, is a painful skin condition classically seen in patients with end-stage renal disease (ESRD), particularly those on chronic dialysis.1,2 It also has increasingly been reported in patients with normal renal function and calcium and phosphate homeostasis.3,4 Effective diagnosis and management of calciphylaxis remains challenging for physicians.2,5 The condition is characterized by tissue ischemia caused by calcification of cutaneous arteriolar vessels. As a result, calciphylaxis is associated with high mortality rates, ranging from 60% to 80%.5,6 Excruciating pain and nonhealing ulcers often lead to recurrent hospitalizations and infectious complications,7 and poor nutritional status, chronic pain, depression, and insomnia can further complicate recovery and lead to poor quality of life.8

We provide an update on calciphylaxis etiopathogenesis, diagnosis, and management. We also highlight some challenges faced in managing this potentially fatal condition.

Epidemiology

Calciphylaxis is considered a rare dermatosis with an estimated annual incidence of 1% to 4% in ESRD patients on dialysis. Recent data suggest that incidence of calciphylaxis is rising,5,7,9 which may stem from an increased use of calcium-based phosphate binders, an actual rise in disease incidence, and/or increased recognition of the disease.5 It is difficult to estimate the exact disease burden of calciphylaxis because the diagnostic criteria are not well defined, often leading to missed or delayed diagnosis.3,10 Furthermore, there is no centralized registry for calciphylaxis cases.3

Etiology and Pathogenesis

Calciphylaxis is thought to have a multifactorial etiology with the exact cause or trigger unknown.7 A long list of risk factors and triggers is associated with the condition (Table 1). Calciphylaxis primarily affects small arteries (40–600 μm in diameter) that become calcified due to an imbalance between inhibitors and promoters of calcification.2,11 Fetuin-A and matrix Gla protein inhibit vascular calcification and are downregulated in calciphylaxis.12,13 Dysfunctional calcium, phosphate, and parathyroid hormone regulatory pathways provide an increased substrate for the process of calcification, which causes endothelial damage and microthrombosis, resulting in tissue ischemia and infarction.14,15 Notably, there is growing interest in the role of vitamin K in the pathogenesis of calciphylaxis. Vitamin K inhibits vascular calcification, possibly by increasing the circulating levels of carboxylated matrix Gla protein.16

Clinical Features

Calciphylaxis is most commonly seen on the legs, abdomen, and buttocks.2 Patients with ESRD commonly develop proximal lesions affecting adipose-rich sites and have a poor prognosis. Distal lesions are more common in patients with nonuremic calciphylaxis, and mortality rates are lower in this population.2

Early lesions present as painful skin nodules or indurated plaques that often are rock-hard or firm to palpation with overlying mottling or a livedoid pattern (Figure, A). Early lesions progress from livedo reticularis to livedo racemosa and then to retiform purpura (Figure, B). Purpuric lesions later evolve into black eschars (Figure, C), then to necrotic, ulcerated, malodorous plaques or nodules in later stages of the disease (Figure, D). Lesions also may develop a gangrenous sclerotic appearance.2,5

Figure
Early lesions of calciphylaxis often appear as indurated plaques with overlying mottling or livedoid pattern (A) that progress to retiform purpura (B). Purpuric lesions then evolve into black eschars (C). In later stages, necrotic, ulcerated, malodorous plaques or nodules are present (D).

Although most patients with calciphylaxis have ESRD, nonuremic patients also can develop the disease. Those with calciphylaxis who do not have renal dysfunction frequently have other risk factors for the disease and often report another notable health problem in the weeks or months prior to presentation.4 More than half of patients with calciphylaxis become bedridden or require use of a wheelchair.17 Pain is characteristically severe throughout the course of the disease; it may even precede the appearance of the skin lesions.18 Because the pain is associated with ischemia, it tends to be relatively refractory to treatment with opioids. Rare extracutaneous vascular calcifications may lead to visual impairment, gastrointestinal tract bleeding, and myopathy.5,9,19,20

Diagnosis

Considering the high morbidity and mortality associated with calciphylaxis, it is important to provide accurate and timely diagnosis; however, there currently are no validated diagnostic criteria for calciphylaxis. Careful correlation of clinical and histologic findings is required. Calciphylaxis biopsies have demonstrated medial calcification and proliferation of the intima of small- to medium-sized arteries.21 Lobular and septal panniculitis and extravascular soft-tissue calcification, particularly stippled calcification of the eccrine sweat glands, also has been seen.2,22 Special calcium stains (eg, von Kossa, Alizarin red) increase the sensitivity of biopsy by highlighting subtle areas of intravascular and extravascular calcification.5,23 Sufficient sampling of subcutaneous tissue and specimen evaluation by an experienced dermatopathologist are necessary to ensure proper interpretation of the histologic findings.

Despite these measures, skin biopsies may be nondiagnostic or falsely negative; therefore, when there is high clinical suspicion, it may be appropriate to move forward with a presumptive diagnosis of calciphylaxis even if the histologic findings are nondiagnostic.1,9,24 It also is worth noting that localized progression and ulceration may occur following skin biopsy, such that biopsy may even be contraindicated in certain cases (eg, penile calciphylaxis).

Standard laboratory workup for calciphylaxis includes evaluation for associated risk factors as well as exclusion of other conditions in the differential diagnosis (Table 2). Blood tests to evaluate for risk factors include liver and renal function tests, a complete metabolic panel, parathyroid hormone level, and serum albumin level.5 Elevated calcium and phosphate levels may signal disturbed calcium and phosphate homeostasis but are neither sensitive nor specific for the diagnosis.25 Complete blood cell count, blood cultures, thorough hypercoagulability workup (including but not limited to antiphospholipid antibodies, proteins C and S, factor V Leiden, antithrombin III, homocysteine, methylenetetrahydrofolate reductase mutation, and cryoglobulins), rheumatoid factor, antineutrophil cytoplasmic antibodies, and antinuclear antibody testing may be relevant to help identify contributing factors or mimickers of calciphylaxis.5 Various imaging modalities also have been used to evaluate for the presence of soft-tissue calcification in areas of suspected calciphylaxis, including radiography, mammography, computed tomography, ultrasonography, nuclear bone scintigraphy, and spectroscopy.2,26,27 Unfortunately, there currently is no standardized reproducible imaging modality for reliable diagnosis of calciphylaxis. Ultimately, histologic and radiographic findings should always be interpreted in the context of relevant clinical findings.2,9

 

 

Prevention

Reduction of the net calcium phosphorus product may help reduce the risk of calciphylaxis in ESRD patients, which can be accomplished by using non–calcium-phosphate binders, adequate dialysis, and restricting use of vitamin D and vitamin K antagonists.2,5 There are limited data regarding the benefits of using bisphosphonates and cinacalcet in ESRD patients on dialysis to prevent calciphylaxis.28,29

Management

Management of calciphylaxis is multifactorial. Besides dermatology and nephrology, specialists in pain management, wound care, plastic surgery, and nutrition are critical partners in management.1,5,9,30 Nephrologists can help optimize calcium and phosphate balance and ensure adequate dialysis. Pain specialists can aid in creating aggressive multiagent pain regimens that target the neuropathic/ischemic and physical aspects of calciphylaxis pain. When appropriate, nutrition specialists can help establish high-protein, low-phosphorus diets, and wound specialists can provide access to advanced wound dressings and adjunctive hyperbaric oxygen therapy. Plastic surgeons can provide conservative debridement procedures in a subset of patients, usually those with distal stable disease.

The limited understanding of the etiopathogenesis of calciphylaxis and the lack of data on its management are reflected in the limited treatment options for the disease (Table 3).2,5,9 There are no formal algorithms for the treatment of calciphylaxis. Therapeutic trials are scarce, and most of the current treatment recommendations are based on small retrospective reports or case series. Sodium thiosulfate has been the most widely used treatment option since 2004, when its use in calciphylaxis was first reported.31 Sodium thiosulfate chelates calcium and is thought to have antioxidant and vasodilatory properties.32 There are a few promising clinical trials and large-scale studies (Table 4) that aim to evaluate the efficacy of existing treatments (eg, sodium thiosulfate) as well as novel treatment options such as lanthanum carbonate, SNF472 (hexasodium phytate), and vitamin K.33-36

Prognosis

Calciphylaxis is a potentially fatal condition with a poor prognosis and a median survival rate of approximately 1 year following the appearance of skin lesions.37-39 Patients with proximal lesions and those on peritoneal dialysis (as opposed to hemodialysis) have a worse prognosis.40 Mortality rates are estimated to be 30% at 6 months, 50% at 12 months, and 80% at 2 years, with sepsis secondary to infection of cutaneous ulcers being the leading cause of death.37-39 The impact of calciphylaxis on patient quality of life and activities of daily living is severe.8,17

Future Directions

Multi-institution cohort studies and collaborative registries are needed to provide updated information related to the epidemiology, diagnosis, treatment, morbidity, and mortality associated with calciphylaxis and to help formulate evidence-based diagnostic criteria. Radiographic and histologic studies, as well as other tools for early and accurate diagnosis of calciphylaxis, should be studied for feasibility, accuracy, and reproducibility. The incidence of nonuremic calciphylaxis points toward pathogenic pathways besides those based on the bone-mineral axis. Basic science research directed at improving understanding of the pathophysiology of calciphylaxis would be helpful in devising new treatment strategies targeting these pathways. Establishment of a collaborative, multi-institutional calciphylaxis working group would enable experts to formulate therapeutic guidelines based on current evidence. Such a group could facilitate initiation of large prospective studies to establish the efficacy of existing and new treatment modalities for calciphylaxis. A working group within the Society for Dermatology Hospitalists has been tasked with addressing these issues and is currently establishing a multicenter calciphylaxis database.

Calciphylaxis, also known as calcific uremic arteriolopathy, is a painful skin condition classically seen in patients with end-stage renal disease (ESRD), particularly those on chronic dialysis.1,2 It also has increasingly been reported in patients with normal renal function and calcium and phosphate homeostasis.3,4 Effective diagnosis and management of calciphylaxis remains challenging for physicians.2,5 The condition is characterized by tissue ischemia caused by calcification of cutaneous arteriolar vessels. As a result, calciphylaxis is associated with high mortality rates, ranging from 60% to 80%.5,6 Excruciating pain and nonhealing ulcers often lead to recurrent hospitalizations and infectious complications,7 and poor nutritional status, chronic pain, depression, and insomnia can further complicate recovery and lead to poor quality of life.8

We provide an update on calciphylaxis etiopathogenesis, diagnosis, and management. We also highlight some challenges faced in managing this potentially fatal condition.

Epidemiology

Calciphylaxis is considered a rare dermatosis with an estimated annual incidence of 1% to 4% in ESRD patients on dialysis. Recent data suggest that incidence of calciphylaxis is rising,5,7,9 which may stem from an increased use of calcium-based phosphate binders, an actual rise in disease incidence, and/or increased recognition of the disease.5 It is difficult to estimate the exact disease burden of calciphylaxis because the diagnostic criteria are not well defined, often leading to missed or delayed diagnosis.3,10 Furthermore, there is no centralized registry for calciphylaxis cases.3

Etiology and Pathogenesis

Calciphylaxis is thought to have a multifactorial etiology with the exact cause or trigger unknown.7 A long list of risk factors and triggers is associated with the condition (Table 1). Calciphylaxis primarily affects small arteries (40–600 μm in diameter) that become calcified due to an imbalance between inhibitors and promoters of calcification.2,11 Fetuin-A and matrix Gla protein inhibit vascular calcification and are downregulated in calciphylaxis.12,13 Dysfunctional calcium, phosphate, and parathyroid hormone regulatory pathways provide an increased substrate for the process of calcification, which causes endothelial damage and microthrombosis, resulting in tissue ischemia and infarction.14,15 Notably, there is growing interest in the role of vitamin K in the pathogenesis of calciphylaxis. Vitamin K inhibits vascular calcification, possibly by increasing the circulating levels of carboxylated matrix Gla protein.16

Clinical Features

Calciphylaxis is most commonly seen on the legs, abdomen, and buttocks.2 Patients with ESRD commonly develop proximal lesions affecting adipose-rich sites and have a poor prognosis. Distal lesions are more common in patients with nonuremic calciphylaxis, and mortality rates are lower in this population.2

Early lesions present as painful skin nodules or indurated plaques that often are rock-hard or firm to palpation with overlying mottling or a livedoid pattern (Figure, A). Early lesions progress from livedo reticularis to livedo racemosa and then to retiform purpura (Figure, B). Purpuric lesions later evolve into black eschars (Figure, C), then to necrotic, ulcerated, malodorous plaques or nodules in later stages of the disease (Figure, D). Lesions also may develop a gangrenous sclerotic appearance.2,5

Figure
Early lesions of calciphylaxis often appear as indurated plaques with overlying mottling or livedoid pattern (A) that progress to retiform purpura (B). Purpuric lesions then evolve into black eschars (C). In later stages, necrotic, ulcerated, malodorous plaques or nodules are present (D).

Although most patients with calciphylaxis have ESRD, nonuremic patients also can develop the disease. Those with calciphylaxis who do not have renal dysfunction frequently have other risk factors for the disease and often report another notable health problem in the weeks or months prior to presentation.4 More than half of patients with calciphylaxis become bedridden or require use of a wheelchair.17 Pain is characteristically severe throughout the course of the disease; it may even precede the appearance of the skin lesions.18 Because the pain is associated with ischemia, it tends to be relatively refractory to treatment with opioids. Rare extracutaneous vascular calcifications may lead to visual impairment, gastrointestinal tract bleeding, and myopathy.5,9,19,20

Diagnosis

Considering the high morbidity and mortality associated with calciphylaxis, it is important to provide accurate and timely diagnosis; however, there currently are no validated diagnostic criteria for calciphylaxis. Careful correlation of clinical and histologic findings is required. Calciphylaxis biopsies have demonstrated medial calcification and proliferation of the intima of small- to medium-sized arteries.21 Lobular and septal panniculitis and extravascular soft-tissue calcification, particularly stippled calcification of the eccrine sweat glands, also has been seen.2,22 Special calcium stains (eg, von Kossa, Alizarin red) increase the sensitivity of biopsy by highlighting subtle areas of intravascular and extravascular calcification.5,23 Sufficient sampling of subcutaneous tissue and specimen evaluation by an experienced dermatopathologist are necessary to ensure proper interpretation of the histologic findings.

Despite these measures, skin biopsies may be nondiagnostic or falsely negative; therefore, when there is high clinical suspicion, it may be appropriate to move forward with a presumptive diagnosis of calciphylaxis even if the histologic findings are nondiagnostic.1,9,24 It also is worth noting that localized progression and ulceration may occur following skin biopsy, such that biopsy may even be contraindicated in certain cases (eg, penile calciphylaxis).

Standard laboratory workup for calciphylaxis includes evaluation for associated risk factors as well as exclusion of other conditions in the differential diagnosis (Table 2). Blood tests to evaluate for risk factors include liver and renal function tests, a complete metabolic panel, parathyroid hormone level, and serum albumin level.5 Elevated calcium and phosphate levels may signal disturbed calcium and phosphate homeostasis but are neither sensitive nor specific for the diagnosis.25 Complete blood cell count, blood cultures, thorough hypercoagulability workup (including but not limited to antiphospholipid antibodies, proteins C and S, factor V Leiden, antithrombin III, homocysteine, methylenetetrahydrofolate reductase mutation, and cryoglobulins), rheumatoid factor, antineutrophil cytoplasmic antibodies, and antinuclear antibody testing may be relevant to help identify contributing factors or mimickers of calciphylaxis.5 Various imaging modalities also have been used to evaluate for the presence of soft-tissue calcification in areas of suspected calciphylaxis, including radiography, mammography, computed tomography, ultrasonography, nuclear bone scintigraphy, and spectroscopy.2,26,27 Unfortunately, there currently is no standardized reproducible imaging modality for reliable diagnosis of calciphylaxis. Ultimately, histologic and radiographic findings should always be interpreted in the context of relevant clinical findings.2,9

 

 

Prevention

Reduction of the net calcium phosphorus product may help reduce the risk of calciphylaxis in ESRD patients, which can be accomplished by using non–calcium-phosphate binders, adequate dialysis, and restricting use of vitamin D and vitamin K antagonists.2,5 There are limited data regarding the benefits of using bisphosphonates and cinacalcet in ESRD patients on dialysis to prevent calciphylaxis.28,29

Management

Management of calciphylaxis is multifactorial. Besides dermatology and nephrology, specialists in pain management, wound care, plastic surgery, and nutrition are critical partners in management.1,5,9,30 Nephrologists can help optimize calcium and phosphate balance and ensure adequate dialysis. Pain specialists can aid in creating aggressive multiagent pain regimens that target the neuropathic/ischemic and physical aspects of calciphylaxis pain. When appropriate, nutrition specialists can help establish high-protein, low-phosphorus diets, and wound specialists can provide access to advanced wound dressings and adjunctive hyperbaric oxygen therapy. Plastic surgeons can provide conservative debridement procedures in a subset of patients, usually those with distal stable disease.

The limited understanding of the etiopathogenesis of calciphylaxis and the lack of data on its management are reflected in the limited treatment options for the disease (Table 3).2,5,9 There are no formal algorithms for the treatment of calciphylaxis. Therapeutic trials are scarce, and most of the current treatment recommendations are based on small retrospective reports or case series. Sodium thiosulfate has been the most widely used treatment option since 2004, when its use in calciphylaxis was first reported.31 Sodium thiosulfate chelates calcium and is thought to have antioxidant and vasodilatory properties.32 There are a few promising clinical trials and large-scale studies (Table 4) that aim to evaluate the efficacy of existing treatments (eg, sodium thiosulfate) as well as novel treatment options such as lanthanum carbonate, SNF472 (hexasodium phytate), and vitamin K.33-36

Prognosis

Calciphylaxis is a potentially fatal condition with a poor prognosis and a median survival rate of approximately 1 year following the appearance of skin lesions.37-39 Patients with proximal lesions and those on peritoneal dialysis (as opposed to hemodialysis) have a worse prognosis.40 Mortality rates are estimated to be 30% at 6 months, 50% at 12 months, and 80% at 2 years, with sepsis secondary to infection of cutaneous ulcers being the leading cause of death.37-39 The impact of calciphylaxis on patient quality of life and activities of daily living is severe.8,17

Future Directions

Multi-institution cohort studies and collaborative registries are needed to provide updated information related to the epidemiology, diagnosis, treatment, morbidity, and mortality associated with calciphylaxis and to help formulate evidence-based diagnostic criteria. Radiographic and histologic studies, as well as other tools for early and accurate diagnosis of calciphylaxis, should be studied for feasibility, accuracy, and reproducibility. The incidence of nonuremic calciphylaxis points toward pathogenic pathways besides those based on the bone-mineral axis. Basic science research directed at improving understanding of the pathophysiology of calciphylaxis would be helpful in devising new treatment strategies targeting these pathways. Establishment of a collaborative, multi-institutional calciphylaxis working group would enable experts to formulate therapeutic guidelines based on current evidence. Such a group could facilitate initiation of large prospective studies to establish the efficacy of existing and new treatment modalities for calciphylaxis. A working group within the Society for Dermatology Hospitalists has been tasked with addressing these issues and is currently establishing a multicenter calciphylaxis database.

References
  1. Nigwekar SU, Kroshinsky D, Nazarian RM, et al. Calciphylaxis: risk factors, diagnosis, and treatment. Am J Kidney Dis. 2015;66:133-146.
  2. Nigwekar SU, Thadhani RI, Brandenburg VM. Calciphylaxis. N Engl J Med. 2018;378:1704-1714.
  3. Davis JM. The relationship between obesity and calciphylaxis: a review of the literature. Ostomy Wound Manage. 2016;62:12-18.
  4. Bajaj R, Courbebaisse M, Kroshinsky D, et al. Calciphylaxis in patients with normal renal function: a case series and systematic review. Mayo Clin Proc. 2018;93:1202-1212.
  5. Hafner J, Keusch G, Wahl C, et al. Uremic small-artery disease with medial calcification and intimal hyperplasia (so-called calciphylaxis): a complication of chronic renal failure and benefit from parathyroidectomy. J Am Acad Dermatol. 1995;33:954-962.
  6. Jeong HS, Dominguez AR. Calciphylaxis: controversies in pathogenesis, diagnosis and treatment. Am J Med Sci. 2016;351:217-227.
  7. Westphal SG, Plumb T. Calciphylaxis. In: StatPearls. Treasure Island, FL: StatPearls Publishing; 2018. https://www.ncbi.nlm.nih.gov/books/NBK519020. Accessed November 12, 2018.
  8. Riemer CA, El-Azhary RA, Wu KL, et al. Underreported use of palliative care and patient-reported outcome measures to address reduced quality of life in patients with calciphylaxis: a systematic review. Br J Dermatol. 2017;177:1510-1518.
  9. Nigwekar SU. Calciphylaxis. Curr Opin Nephrol Hypertens. 2017;26:276-281.
  10. Fine A, Fontaine B. Calciphylaxis: the beginning of the end? Perit Dial Int. 2008;28:268-270.
  11. Lin WT, Chao CM. Tumoral calcinosis in renal failure. QJM. 2014;107:387.
  12. Schafer C, Heiss A, Schwarz A, et al. The serum protein alpha 2-Heremans-Schmid glycoprotein/fetuin-A is a systemically acting inhibitor of ectopic calcification. J Clin Invest. 2003;112:357-366.
  13. Luo G, Ducy P, McKee MD, et al. Spontaneous calcification of arteries and cartilage in mice lacking matrix GLA protein. Nature. 1997;386:78-81.
  14. Bleyer AJ, Choi M, Igwemezie B, et al. A case control study of proximal calciphylaxis. Am J Kidney Dis. 1998;32:376-383.
  15. Ahmed S, O’Neill KD, Hood AF, et al. Calciphylaxis is associated with hyperphosphatemia and increased osteopontin expression by vascular smooth muscle cells. Am J Kidney Dis. 2001;37:267-276.
  16. Nigwekar SU, Bloch DB, Nazarian RM, et al. Vitamin K-dependent carboxylation of matrix gla protein influences the risk of calciphylaxis. J Am Soc Nephrol. 2017;28:1717-1722.
  17. Weenig RH, Sewell LD, Davis MD, et al. Calciphylaxis: natural history, risk factor analysis, and outcome. J Am Acad Dermatol. 2007;56:569-579.
  18. Polizzotto MN, Bryan T, Ashby MA, et al. Symptomatic management of calciphylaxis: a case series and review of the literature. J Pain Symptom Manage. 2006;32:186-190.
  19. Gupta N, Haq KF, Mahajan S, et al. Gastrointestinal bleeding secondary to calciphylaxis. Am J Case Rep. 2015;16:818-822.
  20. Edelstein CL, Wickham MK, Kirby PA. Systemic calciphylaxis presenting as a painful, proximal myopathy. Postgrad Med J. 1992;68:209-211.
  21. Mochel MC, Arakari RY, Wang G, et al. Cutaneous calciphylaxis: a retrospective histopathologic evaluation. Am J Dermatopathol. 2013;35:582-586.
  22. Chen TY, Lehman JS, Gibson LE, et al. Histopathology of calciphylaxis: cohort study with clinical correlations. Am J Dermatopathol. 2017;39:795-802.
  23. Cassius C, Moguelet P, Monfort JB, et al. Calciphylaxis in haemodialysed patients: diagnostic value of calcifications in cutaneous biopsy. Br J Dermatol. 2018;178:292-293.
  24. Sreedhar A, Sheikh HA, Scagliotti CJ, et al. Advanced-stage calciphylaxis: think before you punch. Cleve Clin J Med. 2016;83:562-564.
  25. Brandenburg VM, Kramann R, Rothe H, et al. Calcific uraemic arteriolopathy (calciphylaxis): data from a large nation-wide registry. Nephrol Dial Transplant. 2017;32:126-132.
  26. Paul S, Rabito CA, Vedak P, et al. The role of bone scintigraphy in the diagnosis of calciphylaxis. JAMA Dermatol. 2017;153:101-103.
  27. Shmidt E, Murthy NS, Knudsen JM, et al. Net-like pattern of calcification on plain soft-tissue radiographs in patients with calciphylaxis. J Am Acad Dermatol. 2012;67:1296-1301.
  28. EVOLVE Trial Investigators; Chertow GM, Block GA, Correa-Rotter R, et al. Effect of cinacalcet on cardiovascular disease in patients undergoing dialysis. N Engl J Med. 2012;367:2482-2494.
  29. Rogers NM, Teubner DJO, Coates PT. Calcific uremic arteriolopathy: advances in pathogenesis and treatment. Semin Dial. 2007;20:150-157.
  30. Nigwekar SU. Multidisciplinary approach to calcific uremic arteriolopathy. Curr Opin Nephrol Hypertens. 2015;24:531-537.
  31. Cicone JS, Petronis JB, Embert CD, et al. Successful treatment of calciphylaxis with intravenous sodium thiosulfate. Am J Kidney Dis. 2004;43:1104-1108.
  32. Chen NX, O’Neill K, Akl NK, et al. Adipocyte induced arterial calcification is prevented with sodium thiosulfate. Biochem Biophys Res Commun. 2014;449:151-156.
  33. Chan MR, Ghandour F, Murali NS, et al. Pilot study of the effect of lanthanum carbonate in patients with calciphylaxis: a Wisconsin Network for Health Research (WiNHR) study. J Nephrol Ther. 2014;4:1000162.
  34. Perelló J, Gómez M, Ferrer MD, et al. SNF472, a novel inhibitor of vascular calcification, could be administered during hemodialysis to attain potentially therapeutic phytate levels. J Nephrol. 2018;31:287-296.
  35. Christiadi D, Singer RF. Calciphylaxis in a dialysis patient successfully treated with high-dose vitamin K supplementation. Clin Kidney J. 2018;11:528-529.
  36. Caluwe R, Vandecasteele S, Van Vlem B, et al. Vitamin K2 supplementation in haemodialysis patients: a randomized dose-finding study. Nephrol Dial Transplant. 2014;29:1385-1390.
  37. McCarthy JT, El-Azhary RA, Patzelt MT, et al. Survival, risk factors, and effect of treatment in 101 patients with calciphylaxis. Mayo Clin Proc. 2016;91:1384-1394.
  38. Fine A, Zacharias J. Calciphylaxis is usually non-ulcerating: risk factors, outcome and therapy. Kidney Int. 2002;61:2210-2217.
  39. Nigwekar SU, Zhao S, Wenger J, et al. A nationally representative study of calcific uremic arteriolopathy risk factors. J Am Soc Nephrol. 2016;27:3421-3429.
  40. Zhang Y, Corapi KM, Luongo M, et al. Calciphylaxis in peritoneal dialysis patients: a single center cohort study. Int J Nephrol Renovasc Dis. 2016;9:235-241.
References
  1. Nigwekar SU, Kroshinsky D, Nazarian RM, et al. Calciphylaxis: risk factors, diagnosis, and treatment. Am J Kidney Dis. 2015;66:133-146.
  2. Nigwekar SU, Thadhani RI, Brandenburg VM. Calciphylaxis. N Engl J Med. 2018;378:1704-1714.
  3. Davis JM. The relationship between obesity and calciphylaxis: a review of the literature. Ostomy Wound Manage. 2016;62:12-18.
  4. Bajaj R, Courbebaisse M, Kroshinsky D, et al. Calciphylaxis in patients with normal renal function: a case series and systematic review. Mayo Clin Proc. 2018;93:1202-1212.
  5. Hafner J, Keusch G, Wahl C, et al. Uremic small-artery disease with medial calcification and intimal hyperplasia (so-called calciphylaxis): a complication of chronic renal failure and benefit from parathyroidectomy. J Am Acad Dermatol. 1995;33:954-962.
  6. Jeong HS, Dominguez AR. Calciphylaxis: controversies in pathogenesis, diagnosis and treatment. Am J Med Sci. 2016;351:217-227.
  7. Westphal SG, Plumb T. Calciphylaxis. In: StatPearls. Treasure Island, FL: StatPearls Publishing; 2018. https://www.ncbi.nlm.nih.gov/books/NBK519020. Accessed November 12, 2018.
  8. Riemer CA, El-Azhary RA, Wu KL, et al. Underreported use of palliative care and patient-reported outcome measures to address reduced quality of life in patients with calciphylaxis: a systematic review. Br J Dermatol. 2017;177:1510-1518.
  9. Nigwekar SU. Calciphylaxis. Curr Opin Nephrol Hypertens. 2017;26:276-281.
  10. Fine A, Fontaine B. Calciphylaxis: the beginning of the end? Perit Dial Int. 2008;28:268-270.
  11. Lin WT, Chao CM. Tumoral calcinosis in renal failure. QJM. 2014;107:387.
  12. Schafer C, Heiss A, Schwarz A, et al. The serum protein alpha 2-Heremans-Schmid glycoprotein/fetuin-A is a systemically acting inhibitor of ectopic calcification. J Clin Invest. 2003;112:357-366.
  13. Luo G, Ducy P, McKee MD, et al. Spontaneous calcification of arteries and cartilage in mice lacking matrix GLA protein. Nature. 1997;386:78-81.
  14. Bleyer AJ, Choi M, Igwemezie B, et al. A case control study of proximal calciphylaxis. Am J Kidney Dis. 1998;32:376-383.
  15. Ahmed S, O’Neill KD, Hood AF, et al. Calciphylaxis is associated with hyperphosphatemia and increased osteopontin expression by vascular smooth muscle cells. Am J Kidney Dis. 2001;37:267-276.
  16. Nigwekar SU, Bloch DB, Nazarian RM, et al. Vitamin K-dependent carboxylation of matrix gla protein influences the risk of calciphylaxis. J Am Soc Nephrol. 2017;28:1717-1722.
  17. Weenig RH, Sewell LD, Davis MD, et al. Calciphylaxis: natural history, risk factor analysis, and outcome. J Am Acad Dermatol. 2007;56:569-579.
  18. Polizzotto MN, Bryan T, Ashby MA, et al. Symptomatic management of calciphylaxis: a case series and review of the literature. J Pain Symptom Manage. 2006;32:186-190.
  19. Gupta N, Haq KF, Mahajan S, et al. Gastrointestinal bleeding secondary to calciphylaxis. Am J Case Rep. 2015;16:818-822.
  20. Edelstein CL, Wickham MK, Kirby PA. Systemic calciphylaxis presenting as a painful, proximal myopathy. Postgrad Med J. 1992;68:209-211.
  21. Mochel MC, Arakari RY, Wang G, et al. Cutaneous calciphylaxis: a retrospective histopathologic evaluation. Am J Dermatopathol. 2013;35:582-586.
  22. Chen TY, Lehman JS, Gibson LE, et al. Histopathology of calciphylaxis: cohort study with clinical correlations. Am J Dermatopathol. 2017;39:795-802.
  23. Cassius C, Moguelet P, Monfort JB, et al. Calciphylaxis in haemodialysed patients: diagnostic value of calcifications in cutaneous biopsy. Br J Dermatol. 2018;178:292-293.
  24. Sreedhar A, Sheikh HA, Scagliotti CJ, et al. Advanced-stage calciphylaxis: think before you punch. Cleve Clin J Med. 2016;83:562-564.
  25. Brandenburg VM, Kramann R, Rothe H, et al. Calcific uraemic arteriolopathy (calciphylaxis): data from a large nation-wide registry. Nephrol Dial Transplant. 2017;32:126-132.
  26. Paul S, Rabito CA, Vedak P, et al. The role of bone scintigraphy in the diagnosis of calciphylaxis. JAMA Dermatol. 2017;153:101-103.
  27. Shmidt E, Murthy NS, Knudsen JM, et al. Net-like pattern of calcification on plain soft-tissue radiographs in patients with calciphylaxis. J Am Acad Dermatol. 2012;67:1296-1301.
  28. EVOLVE Trial Investigators; Chertow GM, Block GA, Correa-Rotter R, et al. Effect of cinacalcet on cardiovascular disease in patients undergoing dialysis. N Engl J Med. 2012;367:2482-2494.
  29. Rogers NM, Teubner DJO, Coates PT. Calcific uremic arteriolopathy: advances in pathogenesis and treatment. Semin Dial. 2007;20:150-157.
  30. Nigwekar SU. Multidisciplinary approach to calcific uremic arteriolopathy. Curr Opin Nephrol Hypertens. 2015;24:531-537.
  31. Cicone JS, Petronis JB, Embert CD, et al. Successful treatment of calciphylaxis with intravenous sodium thiosulfate. Am J Kidney Dis. 2004;43:1104-1108.
  32. Chen NX, O’Neill K, Akl NK, et al. Adipocyte induced arterial calcification is prevented with sodium thiosulfate. Biochem Biophys Res Commun. 2014;449:151-156.
  33. Chan MR, Ghandour F, Murali NS, et al. Pilot study of the effect of lanthanum carbonate in patients with calciphylaxis: a Wisconsin Network for Health Research (WiNHR) study. J Nephrol Ther. 2014;4:1000162.
  34. Perelló J, Gómez M, Ferrer MD, et al. SNF472, a novel inhibitor of vascular calcification, could be administered during hemodialysis to attain potentially therapeutic phytate levels. J Nephrol. 2018;31:287-296.
  35. Christiadi D, Singer RF. Calciphylaxis in a dialysis patient successfully treated with high-dose vitamin K supplementation. Clin Kidney J. 2018;11:528-529.
  36. Caluwe R, Vandecasteele S, Van Vlem B, et al. Vitamin K2 supplementation in haemodialysis patients: a randomized dose-finding study. Nephrol Dial Transplant. 2014;29:1385-1390.
  37. McCarthy JT, El-Azhary RA, Patzelt MT, et al. Survival, risk factors, and effect of treatment in 101 patients with calciphylaxis. Mayo Clin Proc. 2016;91:1384-1394.
  38. Fine A, Zacharias J. Calciphylaxis is usually non-ulcerating: risk factors, outcome and therapy. Kidney Int. 2002;61:2210-2217.
  39. Nigwekar SU, Zhao S, Wenger J, et al. A nationally representative study of calcific uremic arteriolopathy risk factors. J Am Soc Nephrol. 2016;27:3421-3429.
  40. Zhang Y, Corapi KM, Luongo M, et al. Calciphylaxis in peritoneal dialysis patients: a single center cohort study. Int J Nephrol Renovasc Dis. 2016;9:235-241.
Issue
Cutis - 102(6)
Issue
Cutis - 102(6)
Page Number
395-400
Page Number
395-400
Publications
Publications
Topics
Article Type
Display Headline
Update on Calciphylaxis Etiopathogenesis, Diagnosis, and Management
Display Headline
Update on Calciphylaxis Etiopathogenesis, Diagnosis, and Management
Sections
Inside the Article

Practice Points

  • Maintain a high index of suspicion for calciphylaxis in patients with end-stage renal disease on chronic dialysis presenting with severely painful livedoid plaques or retiform purpura, particularly in fat-rich body sites.
  • Skin biopsies may be limited by biopsy site, inadequate biopsy depth, missed areas of microcalcification, and absence of definitive histologic criteria. Special calcium stains and review by an experienced dermatopathologist may lower the rate of false-negative biopsies.
  • In cases where the most likely clinical diagnosis is calciphylaxis, treatment should be initiated even if definitive histopathology findings are lacking.
  • Treatment should be multimodal, including elimination of risk factors, intravenous sodium thiosulfate, agents addressing calcium-phosphate metabolism, and surgical debridement, if indicated.
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

Short-term lung function better predicts mortality risk in SSc

Article Type
Changed
Thu, 01/31/2019 - 09:04

 

In addition to known risk factors, early reductions in lung function and diffusing capacity increased mortality risk in patients with systemic sclerosis and comorbid interstitial lung disease over 2 years in an analysis of data from the Scleroderma Lung Studies I and II.

Dr. Elizabeth Volkmann, University of California, Los Angeles
Dr. Elizabeth Volkmann

First author Elizabeth R. Volkmann, MD, of the University of California, Los Angeles, and her colleagues also reported that although early treatment with cyclophosphamide or mycophenolate mofetil in these studies led to shorter-term improvement in surrogate measures of outcomes for systemic sclerosis and comorbid interstitial lung disease (SSc-ILD), either medication might not improve long-term outcomes. The findings were reported in the Annals of the Rheumatic Diseases.

The investigators identified specific factors linked to an increased risk of death over a median follow-up of 8 years in 158 systemic sclerosis-interstitial lung disease patients in the Scleroderma Lung Study I and median follow-up of 3.6 years in 142 patients in Scleroderma Lung Study II, both of which were randomized, controlled trials.

After follow-up was complete, 42% of participants in Scleroderma Lung Study I and 21% of participants in Scleroderma Lung Study II died, with the most common cause of death being complications related to systemic sclerosis. In addition, no statistical difference was seen in time to death between the intervention arms of both trials.

In building a risk prediction model based on findings from both trials, the investigators confirmed several known mortality risk predictors in systemic sclerosis, including increased age and baseline skin score, but also found that changes in forced vital capacity and diffusing capacity for carbon monoxide over 2 years were separately linked with a greater risk of death.



“These findings suggest that short-term changes in surrogate measures of systemic sclerosis-interstitial lung disease progression may have important effects on long-term outcomes,” the researchers wrote.

The researchers acknowledged the results emphasize the need to determine optimal length of treatment in patients with systemic sclerosis and comorbid interstitial lung disease.

“Systemic sclerosis providers should closely monitor lung function when interstitial lung disease is present to accurately identify declines in lung function and promptly intervene to improve patient outcomes,” the researchers concluded.

The study was funded by the National Institutes of Health and the Scleroderma Foundation. The authors reported no conflicts of interest.

SOURCE: Volkmann ER et al. Ann Rheum Dis. 2018 Nov 8. doi: 10.1136/annrheumdis-2018-213708.

Publications
Topics
Sections

 

In addition to known risk factors, early reductions in lung function and diffusing capacity increased mortality risk in patients with systemic sclerosis and comorbid interstitial lung disease over 2 years in an analysis of data from the Scleroderma Lung Studies I and II.

Dr. Elizabeth Volkmann, University of California, Los Angeles
Dr. Elizabeth Volkmann

First author Elizabeth R. Volkmann, MD, of the University of California, Los Angeles, and her colleagues also reported that although early treatment with cyclophosphamide or mycophenolate mofetil in these studies led to shorter-term improvement in surrogate measures of outcomes for systemic sclerosis and comorbid interstitial lung disease (SSc-ILD), either medication might not improve long-term outcomes. The findings were reported in the Annals of the Rheumatic Diseases.

The investigators identified specific factors linked to an increased risk of death over a median follow-up of 8 years in 158 systemic sclerosis-interstitial lung disease patients in the Scleroderma Lung Study I and median follow-up of 3.6 years in 142 patients in Scleroderma Lung Study II, both of which were randomized, controlled trials.

After follow-up was complete, 42% of participants in Scleroderma Lung Study I and 21% of participants in Scleroderma Lung Study II died, with the most common cause of death being complications related to systemic sclerosis. In addition, no statistical difference was seen in time to death between the intervention arms of both trials.

In building a risk prediction model based on findings from both trials, the investigators confirmed several known mortality risk predictors in systemic sclerosis, including increased age and baseline skin score, but also found that changes in forced vital capacity and diffusing capacity for carbon monoxide over 2 years were separately linked with a greater risk of death.



“These findings suggest that short-term changes in surrogate measures of systemic sclerosis-interstitial lung disease progression may have important effects on long-term outcomes,” the researchers wrote.

The researchers acknowledged the results emphasize the need to determine optimal length of treatment in patients with systemic sclerosis and comorbid interstitial lung disease.

“Systemic sclerosis providers should closely monitor lung function when interstitial lung disease is present to accurately identify declines in lung function and promptly intervene to improve patient outcomes,” the researchers concluded.

The study was funded by the National Institutes of Health and the Scleroderma Foundation. The authors reported no conflicts of interest.

SOURCE: Volkmann ER et al. Ann Rheum Dis. 2018 Nov 8. doi: 10.1136/annrheumdis-2018-213708.

 

In addition to known risk factors, early reductions in lung function and diffusing capacity increased mortality risk in patients with systemic sclerosis and comorbid interstitial lung disease over 2 years in an analysis of data from the Scleroderma Lung Studies I and II.

Dr. Elizabeth Volkmann, University of California, Los Angeles
Dr. Elizabeth Volkmann

First author Elizabeth R. Volkmann, MD, of the University of California, Los Angeles, and her colleagues also reported that although early treatment with cyclophosphamide or mycophenolate mofetil in these studies led to shorter-term improvement in surrogate measures of outcomes for systemic sclerosis and comorbid interstitial lung disease (SSc-ILD), either medication might not improve long-term outcomes. The findings were reported in the Annals of the Rheumatic Diseases.

The investigators identified specific factors linked to an increased risk of death over a median follow-up of 8 years in 158 systemic sclerosis-interstitial lung disease patients in the Scleroderma Lung Study I and median follow-up of 3.6 years in 142 patients in Scleroderma Lung Study II, both of which were randomized, controlled trials.

After follow-up was complete, 42% of participants in Scleroderma Lung Study I and 21% of participants in Scleroderma Lung Study II died, with the most common cause of death being complications related to systemic sclerosis. In addition, no statistical difference was seen in time to death between the intervention arms of both trials.

In building a risk prediction model based on findings from both trials, the investigators confirmed several known mortality risk predictors in systemic sclerosis, including increased age and baseline skin score, but also found that changes in forced vital capacity and diffusing capacity for carbon monoxide over 2 years were separately linked with a greater risk of death.



“These findings suggest that short-term changes in surrogate measures of systemic sclerosis-interstitial lung disease progression may have important effects on long-term outcomes,” the researchers wrote.

The researchers acknowledged the results emphasize the need to determine optimal length of treatment in patients with systemic sclerosis and comorbid interstitial lung disease.

“Systemic sclerosis providers should closely monitor lung function when interstitial lung disease is present to accurately identify declines in lung function and promptly intervene to improve patient outcomes,” the researchers concluded.

The study was funded by the National Institutes of Health and the Scleroderma Foundation. The authors reported no conflicts of interest.

SOURCE: Volkmann ER et al. Ann Rheum Dis. 2018 Nov 8. doi: 10.1136/annrheumdis-2018-213708.

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

FROM ANNALS OF THE RHEUMATIC DISEASES

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
CME ID
189407
Vitals

 

Key clinical point: Reductions in lung function over a period of 2 years improved modeling estimates of mortality risk in patients with systemic sclerosis-interstitial lung disease.

Major finding: Declines in forced vital capacity and diffusing capacity for carbon monoxide were significant, independent predictors of death over the study duration.

Study details: A long-term follow-up analysis of two independent randomized, controlled trials, which consisted of 158 and 142 adults, respectively.

Disclosures: The study was funded by the National Institutes of Health and the Scleroderma Foundation. The authors reported no conflicts of interest.

Source: Volkmann ER et al. Ann Rheum Dis. 2018 Nov 8. doi: 10.1136/annrheumdis-2018-213708.

Disqus Comments
Default
Use ProPublica

Optimizing Topical Therapy for Onychomycosis: The Importance of Patient Education

Article Type
Changed
Thu, 03/28/2019 - 14:32
Display Headline
Optimizing Topical Therapy for Onychomycosis: The Importance of Patient Education

Onychomycosis is a fungal infection of the nail unit due to dermatophytes, yeasts, and nondermatophyte molds (NDMs). It accounts for approximately 50% of all nail disorders seen in clinical practice and is estimated to affect 10% to 12% of the US population.1,2 Oral medications approved by the US Food and Drug Administration (FDA) include terbinafine and itraconazole, which have demonstrated good efficacy in treating onychomycosis but are associated with potential drug-drug interactions and systemic side effects.3,4 Although liver failure associated with these drugs is rare,5 many patients are anxious about systemic adverse events and therefore prefer to use topical therapies for onychomycosis.

Many patients desire topical therapy but not every patient is an appropriate candidate. Patients who will likely respond well to topical therapy include those with superficial onychomycosis, distal lateral subungual onychomycosis that involves less than 50% of the nail plate surface area (without matrix involvement and a nail plate thickness less than 2 mm), and only up to 3 or 4 nails affected.6 In patients who have contraindications to oral therapy, topical therapy may be the only treatment option. To maximize efficacy of FDA-approved topical agents for onychomycosis therapy, patient education is of utmost importance. Failure to properly counsel the patient on proper medication application may result in decreased antifungal efficacy; poor patient compliance due to lack of improvement; and progression of disease, leading to increased onychodystrophy and pain.

Before initiating therapy, patients should be counseled that treatment with topical drugs is long, requiring daily application of the medication for 6 months for fingernails and 12 months for toenails, based on average nail growth rates (2–3 mm per month for fingernails; 1 mm per month for toenails).7 Patients also are advised to avoid nail polish application during the course of therapy, as clinical trials were performed without nail polish and the true efficacy with nail polish is unknown.8-10 Because patients who have had onychomycosis for shorter durations generally have better cure rates than those who have disease for longer durations, it is prudent to initiate topical therapy as early as possible.11,12 Treating the feet with an antifungal while treating the nails for onychomycosis further enhances efficacy.13,14 There are 3 FDA-approved topical therapies for onychomycosis: ciclopirox nail lacquer 8%, efinaconazole solution 10%, and tavaborole solution 5%.15-17

Ciclopirox is a hydroxypyridone with broad-spectrum antimicrobial activity against dermatophytes, NDMs, yeasts, and bacteria. Its mechanism of action is to chelate polyvalent cations, such as Fe3+, and inhibit fungal metal-dependent enzymes responsible for the degradation of toxic metabolites.15 Ciclopirox nail lacquer 8% was FDA approved for the treatment of onychomycosis in 1999, making it the first topical approved for this purpose. Its indication is for immunocompetent patients with mild to moderate onychomycosis (Trichophyton rubrum) without lunula involvement, with mycologic cure rates of 29% to 36% and complete cure rates of 5.5% to 8.5% (toenails).15 It is the only FDA-approved topical treatment for both fingernails and toenails. Using a brush applicator, it is applied daily to the nail plate and its undersurface, hyponychium, and 5 mm of the surrounding skin. It is important to counsel the patient to remove the lacquer from the nail plate weekly because failure to do so will result in accumulation of numerous layers of medication, such that the active drug cannot reach the site of infection (Figures 1 and 2). The nail plate also should be trimmed and filed weekly by the patient, with monthly clipping/debridement by a physician recommended.6,15

Figure 1. Appearance of the great toenail after application of ciclopirox nail lacquer 8% daily for 6 months without weekly removal of the lacquer. The great toenail shows accumulation of multiple layers of medication, preventing penetration of the active drug to the site of infection. Prior to presenting to our office, the patient was not counselled on proper application.

Figure 2. After vigorous rubbing of the great toenail with alcohol and clipping the lacquer accumulation, the numerous layers of medication were removed from the nail plate. The patient was then counseled on proper medication application.

Efinaconazole is a triazole with antifungal activity against dermatophytes, NDMs, and Candida species. Its mechanism of action is inhibition of lanosterol 14α-demethylase, an enzyme involved in the biosynthesis of ergosterol, which is a component of the fungal cell membrane. Efinaconazole solution 10% was FDA approved in June 2014 for the treatment of toenail onychomycosis due to T rubrum and Trichophyton mentagrophytes, with package insert mycologic cure rates of 53.4% to 55.2% and complete cure rates of 15.2% to 17.8%.9,16 It is applied with a brush applicator to the nail plate, as well as its undersurface, nail folds, and hyponychium. Two drops are recommended for the great toenail and one drop for all other toenails, and no removal of the solution or debridement is required.6,16

Tavaborole is a benzoxaborole with antifungal activity against dermatophytes, NDMs, and yeasts. Its mechanism of action is inhibition of fungal aminoacyl transfer RNA synthetase, thus impeding protein synthesis.18 Tavaborole solution 5% was FDA approved in July 2014 for the treatment of toenail onychomycosis due to T rubrum and T mentagrophytes, with mycologic cure rates of 31.1% and 35.9% and complete cure rates of 6.5% and 9.1%, respectively.11,17 It is applied with a glass pointed-tip dropper to the nail plate, such that the entire nail is covered as well as under the nail tip. No removal of the solution or debridement is required.17

Topical therapies for onychomycosis require long treatment durations, thus excellent compliance and adherence to the treatment protocol are vital to maximize efficacy. Dermatologists who prescribe ciclopirox nail lacquer 8% should counsel patients to remove the lacquer with alcohol weekly, such that the antifungal penetrates the nail plate to reach the site of infection. Monthly debridement also must be clarified before initiating therapy. With all topical therapy for onychomycosis, it is important to treat early, treat concurrently for tinea pedis, and avoid use of nail polish so that patients have the best possible cure rates.

References
  1. Lipner SR, Scher RK. Onychomycosis: diagnosis and therapy. In: Razzaghi-Abyaneh M, Shams-Ghahfarokhi M, Rai M, eds. Medical Mycology: Current Trends and Future Prospects. Boca Raton, FL: CRC Press; 2015:28.
  2. Scher RK, Rich P, Pariser D, et al. The epidemiology, etiology, and pathophysiology of onychomycosis. Semin Cutan Med Surg. 2013;32(2 suppl 1):S2-S4.
  3. Lamisil [package insert]. East Hanover, NJ: Novartis Pharmaceuticals Corporation; 1997.
  4. Sporanox [package insert]. Titusville, NJ: Janssen Pharmaceuticals, Inc; 2001.
  5. National Institutes of Health. Terbinafine. LiverTox website. https://livertox.nlm.nih.gov/Terbinafine.htm. Accessed November 7, 2018.
  6. Lipner SR, Scher RK. Onychomycosis: topical therapy and devices. In: Rubin AI, Jellinek NJ, Daniel CR III, et al, eds. Scher and Daniel’s Nails: Diagnosis, Surgery, Therapy. 4th ed. Cham, Switzerland: Springer International Publishing; 2018:173-184.
  7. Lipner SR, Scher RK. Nail growth evaluation and factors affecting nail growth. In: Humbert P, Fanian F, Maibach HI, et al, eds. Agache’s Measuring the Skin. 2nd ed. Berlin, Germany: Springer; 2017:867-881.
  8. Gupta AK, Elewski BE, Sugarman JL, et al. The efficacy and safety of efinaconazole 10% solution for treatment of mild to moderate onychomycosis: a pooled analysis of two phase 3 randomized trials. J Drugs Dermatol. 2014;13:815-820.
  9. Elewski BE, Rich P, Pollak R, et al. Efinaconazole 10% solution in the treatment of toenail onychomycosis: two phase III multicenter, randomized, double-blind studies. J Am Acad Dermatol. 2013;68:600-608.
  10. Elewski BE, Aly R, Baldwin SL, et al. Efficacy and safety of tavaborole topical solution, 5%, a novel boron-based antifungal agent, for the treatment of toenail onychomycosis: results from 2 randomized phase-III studies. J Am Acad Dermatol. 2015;73:62-69.
  11. Rich P. Efinaconazole topical solution, 10%: the benefits of treating onychomycosis early. J Drugs Dermatol. 2015;14:58-62.
  12. Lipner SR, Scher RK. Efinaconazole 10% topical solution for the topical treatment of onychomycosis of the toenail. Expert Rev Clin Pharmacol. 2015;8:719-731.
  13. Del Rosso JQ. Onychomycosis of toenails and post-hoc analyses with efinaconazole 10% solution once-daily treatment: impact of disease severity and other concomitant associated factors on selection of therapy and therapeutic outcomes. J Clin Aesthet Dermatol. 2016;9:42.
  14. Lipner SR, Scher RK. Management of onychomycosis and co-existing tinea pedis. J Drugs Dermatol. 2015;14:492-494.
  15. Penlac [package insert]. Berwyn, PA: Dermik Laboratories; 2004.
  16. Jublia [package insert]. Bridgewater, NJ: Valeant Pharmaceuticals, LLC; 2014.
  17. Kerydin [package insert]. Palo Alto, CA: Anacor Pharmaceuticals, Inc; 2014.
  18. Rock FL, Mao W, Yaremchuk A, et al. An antifungal agent inhibits an aminoacyl-tRNA synthetase by trapping tRNA in the editing site. Science. 2007;316:1759-1761.
Article PDF
Author and Disclosure Information

Dr. Lipner is from Weill Cornell Medicine, New York, New York. Mr. Ko is from Duke University School of Medicine, Durham, North Carolina.

The authors report no conflict of interest.

Correspondence: Shari R. Lipner, MD, PhD, 1305 York Ave, New York, NY 10021 (shl9032@med.cornell.edu).

Issue
Cutis - 102(6)
Publications
Topics
Page Number
389-390
Sections
Author and Disclosure Information

Dr. Lipner is from Weill Cornell Medicine, New York, New York. Mr. Ko is from Duke University School of Medicine, Durham, North Carolina.

The authors report no conflict of interest.

Correspondence: Shari R. Lipner, MD, PhD, 1305 York Ave, New York, NY 10021 (shl9032@med.cornell.edu).

Author and Disclosure Information

Dr. Lipner is from Weill Cornell Medicine, New York, New York. Mr. Ko is from Duke University School of Medicine, Durham, North Carolina.

The authors report no conflict of interest.

Correspondence: Shari R. Lipner, MD, PhD, 1305 York Ave, New York, NY 10021 (shl9032@med.cornell.edu).

Article PDF
Article PDF

Onychomycosis is a fungal infection of the nail unit due to dermatophytes, yeasts, and nondermatophyte molds (NDMs). It accounts for approximately 50% of all nail disorders seen in clinical practice and is estimated to affect 10% to 12% of the US population.1,2 Oral medications approved by the US Food and Drug Administration (FDA) include terbinafine and itraconazole, which have demonstrated good efficacy in treating onychomycosis but are associated with potential drug-drug interactions and systemic side effects.3,4 Although liver failure associated with these drugs is rare,5 many patients are anxious about systemic adverse events and therefore prefer to use topical therapies for onychomycosis.

Many patients desire topical therapy but not every patient is an appropriate candidate. Patients who will likely respond well to topical therapy include those with superficial onychomycosis, distal lateral subungual onychomycosis that involves less than 50% of the nail plate surface area (without matrix involvement and a nail plate thickness less than 2 mm), and only up to 3 or 4 nails affected.6 In patients who have contraindications to oral therapy, topical therapy may be the only treatment option. To maximize efficacy of FDA-approved topical agents for onychomycosis therapy, patient education is of utmost importance. Failure to properly counsel the patient on proper medication application may result in decreased antifungal efficacy; poor patient compliance due to lack of improvement; and progression of disease, leading to increased onychodystrophy and pain.

Before initiating therapy, patients should be counseled that treatment with topical drugs is long, requiring daily application of the medication for 6 months for fingernails and 12 months for toenails, based on average nail growth rates (2–3 mm per month for fingernails; 1 mm per month for toenails).7 Patients also are advised to avoid nail polish application during the course of therapy, as clinical trials were performed without nail polish and the true efficacy with nail polish is unknown.8-10 Because patients who have had onychomycosis for shorter durations generally have better cure rates than those who have disease for longer durations, it is prudent to initiate topical therapy as early as possible.11,12 Treating the feet with an antifungal while treating the nails for onychomycosis further enhances efficacy.13,14 There are 3 FDA-approved topical therapies for onychomycosis: ciclopirox nail lacquer 8%, efinaconazole solution 10%, and tavaborole solution 5%.15-17

Ciclopirox is a hydroxypyridone with broad-spectrum antimicrobial activity against dermatophytes, NDMs, yeasts, and bacteria. Its mechanism of action is to chelate polyvalent cations, such as Fe3+, and inhibit fungal metal-dependent enzymes responsible for the degradation of toxic metabolites.15 Ciclopirox nail lacquer 8% was FDA approved for the treatment of onychomycosis in 1999, making it the first topical approved for this purpose. Its indication is for immunocompetent patients with mild to moderate onychomycosis (Trichophyton rubrum) without lunula involvement, with mycologic cure rates of 29% to 36% and complete cure rates of 5.5% to 8.5% (toenails).15 It is the only FDA-approved topical treatment for both fingernails and toenails. Using a brush applicator, it is applied daily to the nail plate and its undersurface, hyponychium, and 5 mm of the surrounding skin. It is important to counsel the patient to remove the lacquer from the nail plate weekly because failure to do so will result in accumulation of numerous layers of medication, such that the active drug cannot reach the site of infection (Figures 1 and 2). The nail plate also should be trimmed and filed weekly by the patient, with monthly clipping/debridement by a physician recommended.6,15

Figure 1. Appearance of the great toenail after application of ciclopirox nail lacquer 8% daily for 6 months without weekly removal of the lacquer. The great toenail shows accumulation of multiple layers of medication, preventing penetration of the active drug to the site of infection. Prior to presenting to our office, the patient was not counselled on proper application.

Figure 2. After vigorous rubbing of the great toenail with alcohol and clipping the lacquer accumulation, the numerous layers of medication were removed from the nail plate. The patient was then counseled on proper medication application.

Efinaconazole is a triazole with antifungal activity against dermatophytes, NDMs, and Candida species. Its mechanism of action is inhibition of lanosterol 14α-demethylase, an enzyme involved in the biosynthesis of ergosterol, which is a component of the fungal cell membrane. Efinaconazole solution 10% was FDA approved in June 2014 for the treatment of toenail onychomycosis due to T rubrum and Trichophyton mentagrophytes, with package insert mycologic cure rates of 53.4% to 55.2% and complete cure rates of 15.2% to 17.8%.9,16 It is applied with a brush applicator to the nail plate, as well as its undersurface, nail folds, and hyponychium. Two drops are recommended for the great toenail and one drop for all other toenails, and no removal of the solution or debridement is required.6,16

Tavaborole is a benzoxaborole with antifungal activity against dermatophytes, NDMs, and yeasts. Its mechanism of action is inhibition of fungal aminoacyl transfer RNA synthetase, thus impeding protein synthesis.18 Tavaborole solution 5% was FDA approved in July 2014 for the treatment of toenail onychomycosis due to T rubrum and T mentagrophytes, with mycologic cure rates of 31.1% and 35.9% and complete cure rates of 6.5% and 9.1%, respectively.11,17 It is applied with a glass pointed-tip dropper to the nail plate, such that the entire nail is covered as well as under the nail tip. No removal of the solution or debridement is required.17

Topical therapies for onychomycosis require long treatment durations, thus excellent compliance and adherence to the treatment protocol are vital to maximize efficacy. Dermatologists who prescribe ciclopirox nail lacquer 8% should counsel patients to remove the lacquer with alcohol weekly, such that the antifungal penetrates the nail plate to reach the site of infection. Monthly debridement also must be clarified before initiating therapy. With all topical therapy for onychomycosis, it is important to treat early, treat concurrently for tinea pedis, and avoid use of nail polish so that patients have the best possible cure rates.

Onychomycosis is a fungal infection of the nail unit due to dermatophytes, yeasts, and nondermatophyte molds (NDMs). It accounts for approximately 50% of all nail disorders seen in clinical practice and is estimated to affect 10% to 12% of the US population.1,2 Oral medications approved by the US Food and Drug Administration (FDA) include terbinafine and itraconazole, which have demonstrated good efficacy in treating onychomycosis but are associated with potential drug-drug interactions and systemic side effects.3,4 Although liver failure associated with these drugs is rare,5 many patients are anxious about systemic adverse events and therefore prefer to use topical therapies for onychomycosis.

Many patients desire topical therapy but not every patient is an appropriate candidate. Patients who will likely respond well to topical therapy include those with superficial onychomycosis, distal lateral subungual onychomycosis that involves less than 50% of the nail plate surface area (without matrix involvement and a nail plate thickness less than 2 mm), and only up to 3 or 4 nails affected.6 In patients who have contraindications to oral therapy, topical therapy may be the only treatment option. To maximize efficacy of FDA-approved topical agents for onychomycosis therapy, patient education is of utmost importance. Failure to properly counsel the patient on proper medication application may result in decreased antifungal efficacy; poor patient compliance due to lack of improvement; and progression of disease, leading to increased onychodystrophy and pain.

Before initiating therapy, patients should be counseled that treatment with topical drugs is long, requiring daily application of the medication for 6 months for fingernails and 12 months for toenails, based on average nail growth rates (2–3 mm per month for fingernails; 1 mm per month for toenails).7 Patients also are advised to avoid nail polish application during the course of therapy, as clinical trials were performed without nail polish and the true efficacy with nail polish is unknown.8-10 Because patients who have had onychomycosis for shorter durations generally have better cure rates than those who have disease for longer durations, it is prudent to initiate topical therapy as early as possible.11,12 Treating the feet with an antifungal while treating the nails for onychomycosis further enhances efficacy.13,14 There are 3 FDA-approved topical therapies for onychomycosis: ciclopirox nail lacquer 8%, efinaconazole solution 10%, and tavaborole solution 5%.15-17

Ciclopirox is a hydroxypyridone with broad-spectrum antimicrobial activity against dermatophytes, NDMs, yeasts, and bacteria. Its mechanism of action is to chelate polyvalent cations, such as Fe3+, and inhibit fungal metal-dependent enzymes responsible for the degradation of toxic metabolites.15 Ciclopirox nail lacquer 8% was FDA approved for the treatment of onychomycosis in 1999, making it the first topical approved for this purpose. Its indication is for immunocompetent patients with mild to moderate onychomycosis (Trichophyton rubrum) without lunula involvement, with mycologic cure rates of 29% to 36% and complete cure rates of 5.5% to 8.5% (toenails).15 It is the only FDA-approved topical treatment for both fingernails and toenails. Using a brush applicator, it is applied daily to the nail plate and its undersurface, hyponychium, and 5 mm of the surrounding skin. It is important to counsel the patient to remove the lacquer from the nail plate weekly because failure to do so will result in accumulation of numerous layers of medication, such that the active drug cannot reach the site of infection (Figures 1 and 2). The nail plate also should be trimmed and filed weekly by the patient, with monthly clipping/debridement by a physician recommended.6,15

Figure 1. Appearance of the great toenail after application of ciclopirox nail lacquer 8% daily for 6 months without weekly removal of the lacquer. The great toenail shows accumulation of multiple layers of medication, preventing penetration of the active drug to the site of infection. Prior to presenting to our office, the patient was not counselled on proper application.

Figure 2. After vigorous rubbing of the great toenail with alcohol and clipping the lacquer accumulation, the numerous layers of medication were removed from the nail plate. The patient was then counseled on proper medication application.

Efinaconazole is a triazole with antifungal activity against dermatophytes, NDMs, and Candida species. Its mechanism of action is inhibition of lanosterol 14α-demethylase, an enzyme involved in the biosynthesis of ergosterol, which is a component of the fungal cell membrane. Efinaconazole solution 10% was FDA approved in June 2014 for the treatment of toenail onychomycosis due to T rubrum and Trichophyton mentagrophytes, with package insert mycologic cure rates of 53.4% to 55.2% and complete cure rates of 15.2% to 17.8%.9,16 It is applied with a brush applicator to the nail plate, as well as its undersurface, nail folds, and hyponychium. Two drops are recommended for the great toenail and one drop for all other toenails, and no removal of the solution or debridement is required.6,16

Tavaborole is a benzoxaborole with antifungal activity against dermatophytes, NDMs, and yeasts. Its mechanism of action is inhibition of fungal aminoacyl transfer RNA synthetase, thus impeding protein synthesis.18 Tavaborole solution 5% was FDA approved in July 2014 for the treatment of toenail onychomycosis due to T rubrum and T mentagrophytes, with mycologic cure rates of 31.1% and 35.9% and complete cure rates of 6.5% and 9.1%, respectively.11,17 It is applied with a glass pointed-tip dropper to the nail plate, such that the entire nail is covered as well as under the nail tip. No removal of the solution or debridement is required.17

Topical therapies for onychomycosis require long treatment durations, thus excellent compliance and adherence to the treatment protocol are vital to maximize efficacy. Dermatologists who prescribe ciclopirox nail lacquer 8% should counsel patients to remove the lacquer with alcohol weekly, such that the antifungal penetrates the nail plate to reach the site of infection. Monthly debridement also must be clarified before initiating therapy. With all topical therapy for onychomycosis, it is important to treat early, treat concurrently for tinea pedis, and avoid use of nail polish so that patients have the best possible cure rates.

References
  1. Lipner SR, Scher RK. Onychomycosis: diagnosis and therapy. In: Razzaghi-Abyaneh M, Shams-Ghahfarokhi M, Rai M, eds. Medical Mycology: Current Trends and Future Prospects. Boca Raton, FL: CRC Press; 2015:28.
  2. Scher RK, Rich P, Pariser D, et al. The epidemiology, etiology, and pathophysiology of onychomycosis. Semin Cutan Med Surg. 2013;32(2 suppl 1):S2-S4.
  3. Lamisil [package insert]. East Hanover, NJ: Novartis Pharmaceuticals Corporation; 1997.
  4. Sporanox [package insert]. Titusville, NJ: Janssen Pharmaceuticals, Inc; 2001.
  5. National Institutes of Health. Terbinafine. LiverTox website. https://livertox.nlm.nih.gov/Terbinafine.htm. Accessed November 7, 2018.
  6. Lipner SR, Scher RK. Onychomycosis: topical therapy and devices. In: Rubin AI, Jellinek NJ, Daniel CR III, et al, eds. Scher and Daniel’s Nails: Diagnosis, Surgery, Therapy. 4th ed. Cham, Switzerland: Springer International Publishing; 2018:173-184.
  7. Lipner SR, Scher RK. Nail growth evaluation and factors affecting nail growth. In: Humbert P, Fanian F, Maibach HI, et al, eds. Agache’s Measuring the Skin. 2nd ed. Berlin, Germany: Springer; 2017:867-881.
  8. Gupta AK, Elewski BE, Sugarman JL, et al. The efficacy and safety of efinaconazole 10% solution for treatment of mild to moderate onychomycosis: a pooled analysis of two phase 3 randomized trials. J Drugs Dermatol. 2014;13:815-820.
  9. Elewski BE, Rich P, Pollak R, et al. Efinaconazole 10% solution in the treatment of toenail onychomycosis: two phase III multicenter, randomized, double-blind studies. J Am Acad Dermatol. 2013;68:600-608.
  10. Elewski BE, Aly R, Baldwin SL, et al. Efficacy and safety of tavaborole topical solution, 5%, a novel boron-based antifungal agent, for the treatment of toenail onychomycosis: results from 2 randomized phase-III studies. J Am Acad Dermatol. 2015;73:62-69.
  11. Rich P. Efinaconazole topical solution, 10%: the benefits of treating onychomycosis early. J Drugs Dermatol. 2015;14:58-62.
  12. Lipner SR, Scher RK. Efinaconazole 10% topical solution for the topical treatment of onychomycosis of the toenail. Expert Rev Clin Pharmacol. 2015;8:719-731.
  13. Del Rosso JQ. Onychomycosis of toenails and post-hoc analyses with efinaconazole 10% solution once-daily treatment: impact of disease severity and other concomitant associated factors on selection of therapy and therapeutic outcomes. J Clin Aesthet Dermatol. 2016;9:42.
  14. Lipner SR, Scher RK. Management of onychomycosis and co-existing tinea pedis. J Drugs Dermatol. 2015;14:492-494.
  15. Penlac [package insert]. Berwyn, PA: Dermik Laboratories; 2004.
  16. Jublia [package insert]. Bridgewater, NJ: Valeant Pharmaceuticals, LLC; 2014.
  17. Kerydin [package insert]. Palo Alto, CA: Anacor Pharmaceuticals, Inc; 2014.
  18. Rock FL, Mao W, Yaremchuk A, et al. An antifungal agent inhibits an aminoacyl-tRNA synthetase by trapping tRNA in the editing site. Science. 2007;316:1759-1761.
References
  1. Lipner SR, Scher RK. Onychomycosis: diagnosis and therapy. In: Razzaghi-Abyaneh M, Shams-Ghahfarokhi M, Rai M, eds. Medical Mycology: Current Trends and Future Prospects. Boca Raton, FL: CRC Press; 2015:28.
  2. Scher RK, Rich P, Pariser D, et al. The epidemiology, etiology, and pathophysiology of onychomycosis. Semin Cutan Med Surg. 2013;32(2 suppl 1):S2-S4.
  3. Lamisil [package insert]. East Hanover, NJ: Novartis Pharmaceuticals Corporation; 1997.
  4. Sporanox [package insert]. Titusville, NJ: Janssen Pharmaceuticals, Inc; 2001.
  5. National Institutes of Health. Terbinafine. LiverTox website. https://livertox.nlm.nih.gov/Terbinafine.htm. Accessed November 7, 2018.
  6. Lipner SR, Scher RK. Onychomycosis: topical therapy and devices. In: Rubin AI, Jellinek NJ, Daniel CR III, et al, eds. Scher and Daniel’s Nails: Diagnosis, Surgery, Therapy. 4th ed. Cham, Switzerland: Springer International Publishing; 2018:173-184.
  7. Lipner SR, Scher RK. Nail growth evaluation and factors affecting nail growth. In: Humbert P, Fanian F, Maibach HI, et al, eds. Agache’s Measuring the Skin. 2nd ed. Berlin, Germany: Springer; 2017:867-881.
  8. Gupta AK, Elewski BE, Sugarman JL, et al. The efficacy and safety of efinaconazole 10% solution for treatment of mild to moderate onychomycosis: a pooled analysis of two phase 3 randomized trials. J Drugs Dermatol. 2014;13:815-820.
  9. Elewski BE, Rich P, Pollak R, et al. Efinaconazole 10% solution in the treatment of toenail onychomycosis: two phase III multicenter, randomized, double-blind studies. J Am Acad Dermatol. 2013;68:600-608.
  10. Elewski BE, Aly R, Baldwin SL, et al. Efficacy and safety of tavaborole topical solution, 5%, a novel boron-based antifungal agent, for the treatment of toenail onychomycosis: results from 2 randomized phase-III studies. J Am Acad Dermatol. 2015;73:62-69.
  11. Rich P. Efinaconazole topical solution, 10%: the benefits of treating onychomycosis early. J Drugs Dermatol. 2015;14:58-62.
  12. Lipner SR, Scher RK. Efinaconazole 10% topical solution for the topical treatment of onychomycosis of the toenail. Expert Rev Clin Pharmacol. 2015;8:719-731.
  13. Del Rosso JQ. Onychomycosis of toenails and post-hoc analyses with efinaconazole 10% solution once-daily treatment: impact of disease severity and other concomitant associated factors on selection of therapy and therapeutic outcomes. J Clin Aesthet Dermatol. 2016;9:42.
  14. Lipner SR, Scher RK. Management of onychomycosis and co-existing tinea pedis. J Drugs Dermatol. 2015;14:492-494.
  15. Penlac [package insert]. Berwyn, PA: Dermik Laboratories; 2004.
  16. Jublia [package insert]. Bridgewater, NJ: Valeant Pharmaceuticals, LLC; 2014.
  17. Kerydin [package insert]. Palo Alto, CA: Anacor Pharmaceuticals, Inc; 2014.
  18. Rock FL, Mao W, Yaremchuk A, et al. An antifungal agent inhibits an aminoacyl-tRNA synthetase by trapping tRNA in the editing site. Science. 2007;316:1759-1761.
Issue
Cutis - 102(6)
Issue
Cutis - 102(6)
Page Number
389-390
Page Number
389-390
Publications
Publications
Topics
Article Type
Display Headline
Optimizing Topical Therapy for Onychomycosis: The Importance of Patient Education
Display Headline
Optimizing Topical Therapy for Onychomycosis: The Importance of Patient Education
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Article PDF Media

Strategies to Reduce Youth Indoor Tanning Injuries

Article Type
Changed
Mon, 04/15/2019 - 11:38
Display Headline
Strategies to Reduce Youth Indoor Tanning Injuries

Perusal of any lifestyle magazine reveals photographs of movie stars with sun-kissed skin. One can imagine their carefree lives afford ample time outdoors, a vast departure from the pasty masses trapped in their office cubicles. Our cultural norms dictate that a glowing look is a sign of health and attractiveness. Light-skinned individuals must receive regular exposure to sunlight to maintain their bronzed color. Over the last century, the indoor tanning industry has expanded to fill the niche created by the ceaseless pursuit of the ideal complexion.

 


Indoor tanning use causes up to 170,000 cases of skin cancer per year worldwide.1 Accumulating sunburns early in life is a leading risk factor for melanoma, the deadliest form of skin cancer. Campaigns to spread awareness about the link between UV radiation and skin cancer are ubiquitous. The US Food and Drug Administration (FDA) recommends against the use of tanning beds by minors, and several states have passed laws restricting their access. However, adolescents continue to engage. White female high school students remain frequenters of this practice, with more than 15% reporting current use of indoor tanning facilities.2 It seems targeted outreach and media campaigns are unsuccessful in influencing their behavior, and new approaches are needed.

Tanning-Related Injuries

Concentrated exposure to UV radiation during indoor tanning sessions carries the potential for immediate harm. Public health campaigns have focused on long-term skin cancer risk while overlooking thousands of injuries that occur annually at tanning salons across the country. The US Consumer Product Safety Commission first noted injuries associated with the largely unregulated tanning industry in 1974.3 In response, the FDA limited radiation levels, required indoor tanning devices to have timers and manual off switches, and mandated the use of protective eyewear. These changes sparked industry backlash due to the cost of compliance. The Indoor Tanning Association (no longer in operation) hired a lobbying firm in 2009 that successfully fought to resist further regulation.3

More than 3000 indoor tanning–related injuries are treated in emergency departments annually.4 White women aged 18 to 24 years who visit tanning salons are most likely to sustain injuries. In one study, severe skin burns accounted for 80% of emergency department visits, while the rest were due to fainting, eye injuries, and infections from unsanitary equipment.Timer malfunctions may play a role in tanning bed injuries, as several injured patients have reported falling asleep while tanning.4 Only 5% of tanning salons in North Carolina complied with FDA-recommended exposure schedules in 2003, suggesting that neglect or deliberate override of safety features also may contribute to injury.5

Challenges in Changing Tanning Behaviors

Use of indoor tanning facilities by adolescents is boosted by their misperceptions of peer engagement. Many teenagers overestimate the number of their peers who tan, which influences their own behavior.6 This phenomenon illustrates the importance of perceived social norms in this demographic group. Motivating adolescents to take actions that violate these norms poses a considerable challenge.

To teenagers, the perceived immediate benefits of indoor tanning far outweigh perceived costs. The immediate benefit of indoor tanning is having attractive skin, which may improve social standing and perceived self-worth. When adolescents weigh costs and benefits at different points in time, the present value of future events is discounted when compared to current events. For example, an immediate loss of $1000 is more impactful than losing $1000 ten years down the road. Adolescents are motivated to succeed in the short-term and may heavily discount future adverse effects such as the risk for developing cancer or premature aging of the skin. Therefore, getting a tan may be the “rational” decision even if there is an increased risk of future skin cancer.7

The addiction theory of tanning seeks to explain why individuals continue to tan despite knowledge of the associated risks. Exposure to UV radiation releases endorphins, producing a natural narcotic effect.8 The relaxing feeling sunbathers experience may lead to a phenomenon similar to addictions to opioids, alcohol, tobacco, and sugar. Behavior change is a process that unfolds over time. The 5 stages are precontemplation, contemplation, preparation, action, and maintenance.9 Education falls on deaf ears when the recipients are not ready to consider change. Individuals who are already thinking about cutting back on tanning fall into the category of contemplators and are the most open to educational techniques.9

 

 

Potential Solutions

Despite the dire long-term consequences of melanoma, warning adolescents of the increased cancer risk from tanning is an ineffective dissuasion strategy.10 Solutions that aim to limit tanning behaviors in this population should instead center on decreasing the present utility of a tan. Emphasis on the risk of immediate injury may be one effective route. The costs of potential damage to current appearance, vision, and overall health are not readily discounted by adolescents. Teens who devote time and money to the pursuit of a golden glow place high value on attractiveness. Such individuals respond best to loss-framed messages that focus on the impact of UV exposure on appearance, not just their health.11 However, appearance-motivated individuals may feel threatened by interventions that aim to reduce their decision freedom and display high reactance, leading them to reassert their freedom by resisting antitanning messages.12 Another strategy is altering media messaging to support a wider swathe of skin tones, reducing the social benefits of a tan. To swing the needle on our cultural norms, this intervention will require an enduring effort with backing from media outlets and celebrities.

Taxes on tanning salons and devices provide a basic economic disincentive to adolescents who typically have limited funds. State cigarette tax increases successfully reduced youth consumption of tobacco in the 1990s.13 A provision of the Patient Protection and Affordable Care Act levied a 10% excise tax on tanning salons with promising early results.14 Further taxation may generate revenue for educational campaigns on the injury risks of tanning. Continued safety improvements that limit user exposure to UV radiation and enforcement of FDA regulations also will decrease injury rates. Minimizing the UV output of tanning beds and designing protective equipment for tanners are 2 potential objectives. Improvement of over-the-counter sunless tanning agents also will provide alternatives to catching rays for adolescents who wish to attain a bronzed complexion.

Final Thoughts

Health care providers must assess a patient’s readiness for change and tailor interventions accordingly. Regardless of the method, new approaches to combat adolescent tanning injuries may reduce health care costs and minimize serious public health concerns for the next generation.

References
  1. Firger J. Indoor tanning injuries send thousands to the ER each year. CBS News. December 16, 2014. https://www.cbsnews.com/news/skin-cancer-burns-indoor-tanning-salon-injuries/. Accessed November 7, 2018.
  2. Guy GP, Berkowitz Z, Everett Jones S, et al. Prevalence of indoor tanning and association with sunburn among youth in the United States. JAMA Dermatol. 2017;153:387-390.
  3. Pulley MK. Government tan lines: examining the reach and effectiveness of federal and state efforts to protect consumers from the dangers of indoor tanning. Pepperdine Law Review. 2009;36:1163-1181.
  4. Guy GP Jr, Watson M, Haileyesus T, et al. Indoor tanning–related injuries treated in a national sample of US hospital emergency departments. JAMA Intern Med. 2015;175:309-311.
  5. Hornung RL, Magee KH, Lee WJ, et al. Tanning facility use: are we exceeding Food and Drug Administration limits? J Am Acad Dermatol. 2003;49:655-661.
  6. Hoerster KD, Mayer JA, Woodruff SI, et al. The influence of parents and peers on adolescent indoor tanning behavior: findings from a multi-city sample. J Am Acad Dermatol. 2007;57:990-997.
  7. Feldman SR, Dempsey JR, Grummer S, et al. Implications of a utility model for ultraviolet exposure behavior. J Am Acad Dermatol. 2001;45:718-722.
  8. Okhovat J, Feldman SR. Tanning: an addiction? The Melanoma Letter. 2013 Winter;31:5-7. https://www.skincancer.org/Media/Default/File/File/SCF_ML_31-3.pdf. Accessed November 11, 2017.
  9. Prochaska JO, DiClemente CC, Norcross JC. In search of how people change. applications to addictive behaviors. Am Psychol. 1992;47:1102-1114.
  10. Baker MK. Preventing Skin Cancer in Adolescent Girls Through Intervention With Their Mothers [dissertation]. Johnson City, TN: East Tennessee State University; 2013.
  11. Thomas K, Hevey D, Pertl M, et al. Appearance matters: the frame and focus of health messages influences beliefs about skin cancer. Br J Health Psychol. 2011;16(pt 2):418-429.
  12. Jones JL, Leary MR. Effects of appearance-based admonitions against sun exposure on tanning intentions in young adults. Health Psychol. 1994;13:86-90.
  13. Carpenter C, Cook PJ. Cigarette taxes and youth smoking: new evidence from national, state, and local youth risk behavior surveys. J Health Econ. 2008;27:287-99.
  14. Ryan E. The ‘tanning tax’ is a public health success story. Health Affairs website. https://www.healthaffairs.org/do/10.1377/hblog20170815.061547/full/. Published August 15, 2017. Accessed November 7, 2018.
Article PDF
Author and Disclosure Information

Mr. Hamid and Dr. Feldman are from the Center for Dermatology Research, Department of Dermatology, Wake Forest School of Medicine, Winston-Salem, North Carolina. Dr. Feldman also is from the Departments of Pathology and Public Health Sciences. Dr. Fleischer is from the Department of Dermatology, University of Cincinnati, Ohio. Dr. Siegel is from the Department of Dermatology, SUNY Downstate Medical Center, Brooklyn, and the Brooklyn VA Hospital, New York.

The authors report no conflict of interest.

Correspondence: Ramiz N. Hamid, MPH, Department of Dermatology, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC 27157-1071 (hamid0615@gmail.com).

Issue
Cutis - 102(6)
Publications
Topics
Page Number
383-384
Sections
Author and Disclosure Information

Mr. Hamid and Dr. Feldman are from the Center for Dermatology Research, Department of Dermatology, Wake Forest School of Medicine, Winston-Salem, North Carolina. Dr. Feldman also is from the Departments of Pathology and Public Health Sciences. Dr. Fleischer is from the Department of Dermatology, University of Cincinnati, Ohio. Dr. Siegel is from the Department of Dermatology, SUNY Downstate Medical Center, Brooklyn, and the Brooklyn VA Hospital, New York.

The authors report no conflict of interest.

Correspondence: Ramiz N. Hamid, MPH, Department of Dermatology, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC 27157-1071 (hamid0615@gmail.com).

Author and Disclosure Information

Mr. Hamid and Dr. Feldman are from the Center for Dermatology Research, Department of Dermatology, Wake Forest School of Medicine, Winston-Salem, North Carolina. Dr. Feldman also is from the Departments of Pathology and Public Health Sciences. Dr. Fleischer is from the Department of Dermatology, University of Cincinnati, Ohio. Dr. Siegel is from the Department of Dermatology, SUNY Downstate Medical Center, Brooklyn, and the Brooklyn VA Hospital, New York.

The authors report no conflict of interest.

Correspondence: Ramiz N. Hamid, MPH, Department of Dermatology, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC 27157-1071 (hamid0615@gmail.com).

Article PDF
Article PDF

Perusal of any lifestyle magazine reveals photographs of movie stars with sun-kissed skin. One can imagine their carefree lives afford ample time outdoors, a vast departure from the pasty masses trapped in their office cubicles. Our cultural norms dictate that a glowing look is a sign of health and attractiveness. Light-skinned individuals must receive regular exposure to sunlight to maintain their bronzed color. Over the last century, the indoor tanning industry has expanded to fill the niche created by the ceaseless pursuit of the ideal complexion.

 


Indoor tanning use causes up to 170,000 cases of skin cancer per year worldwide.1 Accumulating sunburns early in life is a leading risk factor for melanoma, the deadliest form of skin cancer. Campaigns to spread awareness about the link between UV radiation and skin cancer are ubiquitous. The US Food and Drug Administration (FDA) recommends against the use of tanning beds by minors, and several states have passed laws restricting their access. However, adolescents continue to engage. White female high school students remain frequenters of this practice, with more than 15% reporting current use of indoor tanning facilities.2 It seems targeted outreach and media campaigns are unsuccessful in influencing their behavior, and new approaches are needed.

Tanning-Related Injuries

Concentrated exposure to UV radiation during indoor tanning sessions carries the potential for immediate harm. Public health campaigns have focused on long-term skin cancer risk while overlooking thousands of injuries that occur annually at tanning salons across the country. The US Consumer Product Safety Commission first noted injuries associated with the largely unregulated tanning industry in 1974.3 In response, the FDA limited radiation levels, required indoor tanning devices to have timers and manual off switches, and mandated the use of protective eyewear. These changes sparked industry backlash due to the cost of compliance. The Indoor Tanning Association (no longer in operation) hired a lobbying firm in 2009 that successfully fought to resist further regulation.3

More than 3000 indoor tanning–related injuries are treated in emergency departments annually.4 White women aged 18 to 24 years who visit tanning salons are most likely to sustain injuries. In one study, severe skin burns accounted for 80% of emergency department visits, while the rest were due to fainting, eye injuries, and infections from unsanitary equipment.Timer malfunctions may play a role in tanning bed injuries, as several injured patients have reported falling asleep while tanning.4 Only 5% of tanning salons in North Carolina complied with FDA-recommended exposure schedules in 2003, suggesting that neglect or deliberate override of safety features also may contribute to injury.5

Challenges in Changing Tanning Behaviors

Use of indoor tanning facilities by adolescents is boosted by their misperceptions of peer engagement. Many teenagers overestimate the number of their peers who tan, which influences their own behavior.6 This phenomenon illustrates the importance of perceived social norms in this demographic group. Motivating adolescents to take actions that violate these norms poses a considerable challenge.

To teenagers, the perceived immediate benefits of indoor tanning far outweigh perceived costs. The immediate benefit of indoor tanning is having attractive skin, which may improve social standing and perceived self-worth. When adolescents weigh costs and benefits at different points in time, the present value of future events is discounted when compared to current events. For example, an immediate loss of $1000 is more impactful than losing $1000 ten years down the road. Adolescents are motivated to succeed in the short-term and may heavily discount future adverse effects such as the risk for developing cancer or premature aging of the skin. Therefore, getting a tan may be the “rational” decision even if there is an increased risk of future skin cancer.7

The addiction theory of tanning seeks to explain why individuals continue to tan despite knowledge of the associated risks. Exposure to UV radiation releases endorphins, producing a natural narcotic effect.8 The relaxing feeling sunbathers experience may lead to a phenomenon similar to addictions to opioids, alcohol, tobacco, and sugar. Behavior change is a process that unfolds over time. The 5 stages are precontemplation, contemplation, preparation, action, and maintenance.9 Education falls on deaf ears when the recipients are not ready to consider change. Individuals who are already thinking about cutting back on tanning fall into the category of contemplators and are the most open to educational techniques.9

 

 

Potential Solutions

Despite the dire long-term consequences of melanoma, warning adolescents of the increased cancer risk from tanning is an ineffective dissuasion strategy.10 Solutions that aim to limit tanning behaviors in this population should instead center on decreasing the present utility of a tan. Emphasis on the risk of immediate injury may be one effective route. The costs of potential damage to current appearance, vision, and overall health are not readily discounted by adolescents. Teens who devote time and money to the pursuit of a golden glow place high value on attractiveness. Such individuals respond best to loss-framed messages that focus on the impact of UV exposure on appearance, not just their health.11 However, appearance-motivated individuals may feel threatened by interventions that aim to reduce their decision freedom and display high reactance, leading them to reassert their freedom by resisting antitanning messages.12 Another strategy is altering media messaging to support a wider swathe of skin tones, reducing the social benefits of a tan. To swing the needle on our cultural norms, this intervention will require an enduring effort with backing from media outlets and celebrities.

Taxes on tanning salons and devices provide a basic economic disincentive to adolescents who typically have limited funds. State cigarette tax increases successfully reduced youth consumption of tobacco in the 1990s.13 A provision of the Patient Protection and Affordable Care Act levied a 10% excise tax on tanning salons with promising early results.14 Further taxation may generate revenue for educational campaigns on the injury risks of tanning. Continued safety improvements that limit user exposure to UV radiation and enforcement of FDA regulations also will decrease injury rates. Minimizing the UV output of tanning beds and designing protective equipment for tanners are 2 potential objectives. Improvement of over-the-counter sunless tanning agents also will provide alternatives to catching rays for adolescents who wish to attain a bronzed complexion.

Final Thoughts

Health care providers must assess a patient’s readiness for change and tailor interventions accordingly. Regardless of the method, new approaches to combat adolescent tanning injuries may reduce health care costs and minimize serious public health concerns for the next generation.

Perusal of any lifestyle magazine reveals photographs of movie stars with sun-kissed skin. One can imagine their carefree lives afford ample time outdoors, a vast departure from the pasty masses trapped in their office cubicles. Our cultural norms dictate that a glowing look is a sign of health and attractiveness. Light-skinned individuals must receive regular exposure to sunlight to maintain their bronzed color. Over the last century, the indoor tanning industry has expanded to fill the niche created by the ceaseless pursuit of the ideal complexion.

 


Indoor tanning use causes up to 170,000 cases of skin cancer per year worldwide.1 Accumulating sunburns early in life is a leading risk factor for melanoma, the deadliest form of skin cancer. Campaigns to spread awareness about the link between UV radiation and skin cancer are ubiquitous. The US Food and Drug Administration (FDA) recommends against the use of tanning beds by minors, and several states have passed laws restricting their access. However, adolescents continue to engage. White female high school students remain frequenters of this practice, with more than 15% reporting current use of indoor tanning facilities.2 It seems targeted outreach and media campaigns are unsuccessful in influencing their behavior, and new approaches are needed.

Tanning-Related Injuries

Concentrated exposure to UV radiation during indoor tanning sessions carries the potential for immediate harm. Public health campaigns have focused on long-term skin cancer risk while overlooking thousands of injuries that occur annually at tanning salons across the country. The US Consumer Product Safety Commission first noted injuries associated with the largely unregulated tanning industry in 1974.3 In response, the FDA limited radiation levels, required indoor tanning devices to have timers and manual off switches, and mandated the use of protective eyewear. These changes sparked industry backlash due to the cost of compliance. The Indoor Tanning Association (no longer in operation) hired a lobbying firm in 2009 that successfully fought to resist further regulation.3

More than 3000 indoor tanning–related injuries are treated in emergency departments annually.4 White women aged 18 to 24 years who visit tanning salons are most likely to sustain injuries. In one study, severe skin burns accounted for 80% of emergency department visits, while the rest were due to fainting, eye injuries, and infections from unsanitary equipment.Timer malfunctions may play a role in tanning bed injuries, as several injured patients have reported falling asleep while tanning.4 Only 5% of tanning salons in North Carolina complied with FDA-recommended exposure schedules in 2003, suggesting that neglect or deliberate override of safety features also may contribute to injury.5

Challenges in Changing Tanning Behaviors

Use of indoor tanning facilities by adolescents is boosted by their misperceptions of peer engagement. Many teenagers overestimate the number of their peers who tan, which influences their own behavior.6 This phenomenon illustrates the importance of perceived social norms in this demographic group. Motivating adolescents to take actions that violate these norms poses a considerable challenge.

To teenagers, the perceived immediate benefits of indoor tanning far outweigh perceived costs. The immediate benefit of indoor tanning is having attractive skin, which may improve social standing and perceived self-worth. When adolescents weigh costs and benefits at different points in time, the present value of future events is discounted when compared to current events. For example, an immediate loss of $1000 is more impactful than losing $1000 ten years down the road. Adolescents are motivated to succeed in the short-term and may heavily discount future adverse effects such as the risk for developing cancer or premature aging of the skin. Therefore, getting a tan may be the “rational” decision even if there is an increased risk of future skin cancer.7

The addiction theory of tanning seeks to explain why individuals continue to tan despite knowledge of the associated risks. Exposure to UV radiation releases endorphins, producing a natural narcotic effect.8 The relaxing feeling sunbathers experience may lead to a phenomenon similar to addictions to opioids, alcohol, tobacco, and sugar. Behavior change is a process that unfolds over time. The 5 stages are precontemplation, contemplation, preparation, action, and maintenance.9 Education falls on deaf ears when the recipients are not ready to consider change. Individuals who are already thinking about cutting back on tanning fall into the category of contemplators and are the most open to educational techniques.9

 

 

Potential Solutions

Despite the dire long-term consequences of melanoma, warning adolescents of the increased cancer risk from tanning is an ineffective dissuasion strategy.10 Solutions that aim to limit tanning behaviors in this population should instead center on decreasing the present utility of a tan. Emphasis on the risk of immediate injury may be one effective route. The costs of potential damage to current appearance, vision, and overall health are not readily discounted by adolescents. Teens who devote time and money to the pursuit of a golden glow place high value on attractiveness. Such individuals respond best to loss-framed messages that focus on the impact of UV exposure on appearance, not just their health.11 However, appearance-motivated individuals may feel threatened by interventions that aim to reduce their decision freedom and display high reactance, leading them to reassert their freedom by resisting antitanning messages.12 Another strategy is altering media messaging to support a wider swathe of skin tones, reducing the social benefits of a tan. To swing the needle on our cultural norms, this intervention will require an enduring effort with backing from media outlets and celebrities.

Taxes on tanning salons and devices provide a basic economic disincentive to adolescents who typically have limited funds. State cigarette tax increases successfully reduced youth consumption of tobacco in the 1990s.13 A provision of the Patient Protection and Affordable Care Act levied a 10% excise tax on tanning salons with promising early results.14 Further taxation may generate revenue for educational campaigns on the injury risks of tanning. Continued safety improvements that limit user exposure to UV radiation and enforcement of FDA regulations also will decrease injury rates. Minimizing the UV output of tanning beds and designing protective equipment for tanners are 2 potential objectives. Improvement of over-the-counter sunless tanning agents also will provide alternatives to catching rays for adolescents who wish to attain a bronzed complexion.

Final Thoughts

Health care providers must assess a patient’s readiness for change and tailor interventions accordingly. Regardless of the method, new approaches to combat adolescent tanning injuries may reduce health care costs and minimize serious public health concerns for the next generation.

References
  1. Firger J. Indoor tanning injuries send thousands to the ER each year. CBS News. December 16, 2014. https://www.cbsnews.com/news/skin-cancer-burns-indoor-tanning-salon-injuries/. Accessed November 7, 2018.
  2. Guy GP, Berkowitz Z, Everett Jones S, et al. Prevalence of indoor tanning and association with sunburn among youth in the United States. JAMA Dermatol. 2017;153:387-390.
  3. Pulley MK. Government tan lines: examining the reach and effectiveness of federal and state efforts to protect consumers from the dangers of indoor tanning. Pepperdine Law Review. 2009;36:1163-1181.
  4. Guy GP Jr, Watson M, Haileyesus T, et al. Indoor tanning–related injuries treated in a national sample of US hospital emergency departments. JAMA Intern Med. 2015;175:309-311.
  5. Hornung RL, Magee KH, Lee WJ, et al. Tanning facility use: are we exceeding Food and Drug Administration limits? J Am Acad Dermatol. 2003;49:655-661.
  6. Hoerster KD, Mayer JA, Woodruff SI, et al. The influence of parents and peers on adolescent indoor tanning behavior: findings from a multi-city sample. J Am Acad Dermatol. 2007;57:990-997.
  7. Feldman SR, Dempsey JR, Grummer S, et al. Implications of a utility model for ultraviolet exposure behavior. J Am Acad Dermatol. 2001;45:718-722.
  8. Okhovat J, Feldman SR. Tanning: an addiction? The Melanoma Letter. 2013 Winter;31:5-7. https://www.skincancer.org/Media/Default/File/File/SCF_ML_31-3.pdf. Accessed November 11, 2017.
  9. Prochaska JO, DiClemente CC, Norcross JC. In search of how people change. applications to addictive behaviors. Am Psychol. 1992;47:1102-1114.
  10. Baker MK. Preventing Skin Cancer in Adolescent Girls Through Intervention With Their Mothers [dissertation]. Johnson City, TN: East Tennessee State University; 2013.
  11. Thomas K, Hevey D, Pertl M, et al. Appearance matters: the frame and focus of health messages influences beliefs about skin cancer. Br J Health Psychol. 2011;16(pt 2):418-429.
  12. Jones JL, Leary MR. Effects of appearance-based admonitions against sun exposure on tanning intentions in young adults. Health Psychol. 1994;13:86-90.
  13. Carpenter C, Cook PJ. Cigarette taxes and youth smoking: new evidence from national, state, and local youth risk behavior surveys. J Health Econ. 2008;27:287-99.
  14. Ryan E. The ‘tanning tax’ is a public health success story. Health Affairs website. https://www.healthaffairs.org/do/10.1377/hblog20170815.061547/full/. Published August 15, 2017. Accessed November 7, 2018.
References
  1. Firger J. Indoor tanning injuries send thousands to the ER each year. CBS News. December 16, 2014. https://www.cbsnews.com/news/skin-cancer-burns-indoor-tanning-salon-injuries/. Accessed November 7, 2018.
  2. Guy GP, Berkowitz Z, Everett Jones S, et al. Prevalence of indoor tanning and association with sunburn among youth in the United States. JAMA Dermatol. 2017;153:387-390.
  3. Pulley MK. Government tan lines: examining the reach and effectiveness of federal and state efforts to protect consumers from the dangers of indoor tanning. Pepperdine Law Review. 2009;36:1163-1181.
  4. Guy GP Jr, Watson M, Haileyesus T, et al. Indoor tanning–related injuries treated in a national sample of US hospital emergency departments. JAMA Intern Med. 2015;175:309-311.
  5. Hornung RL, Magee KH, Lee WJ, et al. Tanning facility use: are we exceeding Food and Drug Administration limits? J Am Acad Dermatol. 2003;49:655-661.
  6. Hoerster KD, Mayer JA, Woodruff SI, et al. The influence of parents and peers on adolescent indoor tanning behavior: findings from a multi-city sample. J Am Acad Dermatol. 2007;57:990-997.
  7. Feldman SR, Dempsey JR, Grummer S, et al. Implications of a utility model for ultraviolet exposure behavior. J Am Acad Dermatol. 2001;45:718-722.
  8. Okhovat J, Feldman SR. Tanning: an addiction? The Melanoma Letter. 2013 Winter;31:5-7. https://www.skincancer.org/Media/Default/File/File/SCF_ML_31-3.pdf. Accessed November 11, 2017.
  9. Prochaska JO, DiClemente CC, Norcross JC. In search of how people change. applications to addictive behaviors. Am Psychol. 1992;47:1102-1114.
  10. Baker MK. Preventing Skin Cancer in Adolescent Girls Through Intervention With Their Mothers [dissertation]. Johnson City, TN: East Tennessee State University; 2013.
  11. Thomas K, Hevey D, Pertl M, et al. Appearance matters: the frame and focus of health messages influences beliefs about skin cancer. Br J Health Psychol. 2011;16(pt 2):418-429.
  12. Jones JL, Leary MR. Effects of appearance-based admonitions against sun exposure on tanning intentions in young adults. Health Psychol. 1994;13:86-90.
  13. Carpenter C, Cook PJ. Cigarette taxes and youth smoking: new evidence from national, state, and local youth risk behavior surveys. J Health Econ. 2008;27:287-99.
  14. Ryan E. The ‘tanning tax’ is a public health success story. Health Affairs website. https://www.healthaffairs.org/do/10.1377/hblog20170815.061547/full/. Published August 15, 2017. Accessed November 7, 2018.
Issue
Cutis - 102(6)
Issue
Cutis - 102(6)
Page Number
383-384
Page Number
383-384
Publications
Publications
Topics
Article Type
Display Headline
Strategies to Reduce Youth Indoor Tanning Injuries
Display Headline
Strategies to Reduce Youth Indoor Tanning Injuries
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

Fewer hypos with CGM in routine pediatric T1DM care

Article Type
Changed
Tue, 05/03/2022 - 15:16

 

BERLIN – A significant reduction in severe hypoglycemia was seen within the first year of using continuous glucose monitoring in a registry study of more than 3,000 children with type 1 diabetes mellitus.

Prior to continuous glucose monitoring (CGM) use, 3.9% of 3,171 individuals, aged a median of 12 years, had hypoglycemia events requiring external help. After 6 months of using CGM, however, the rate of severe hypoglycemia had fallen to 1.2% (P = .10), which remained at 1.2% at 12 months (P = .002). The event rate (events per 100 patient-years) was 10.6 at baseline, 7.8 at 6 months, and 6.1 at 12 months.

Fewer hypoglycemia events leading to coma or convulsion were seen with CGM over time, with 1.3%, 0.6%, and 0.7% of patients reporting at least one event at baseline, 6 months (P = .08), and 12 months (P = .15), respectively. Corresponding event rates were 2.5, 1.6, and 1.7 per 100 patient-years.

“The use of continuous glucose monitoring systems has increased considerably in the past years in individuals with type 1 diabetes,” noted Julia Hermann, a PhD student at Ulm University (Germany), at the annual meeting of the European Association for the Study of Diabetes. “In Germany, for example, CGM use rose considerably when reimbursement by health insurance for CGM started in the summer of 2016.”

Ms. Hermann noted that there were studies showing improved metabolic control with CGM but that clinical studies were often limited by the population of patients studied, restricting treatment to selected patients who may have been adhering better to the use of CGM because they were in a trial.

The aim of the present study was to assess metabolic control and acute complications associated with CGM use in children during its first year of use in a real-world population. Anonymized patient records from the prospective German-Austrian-Luxembourg diabetes patient follow-up (DPV) registry were used.

The DPV database is a standardized, computed-based registry of more than 500,000 adult and pediatric patients with all types of diabetes. It was established in 1995 and receives data from 471 diabetes clinics in Germany, Austria, Luxembourg, and Switzerland.

For the current analysis patients had to be aged under 18 years, have had type 1 diabetes mellitus for at least 1 year, have data available for the months prior to starting CGM, and have at least 1 year of follow-up. For inclusion there also had to be documented use of CMG for at least half of the observation time.

The reduction of severe hypoglycemia observed was not accompanied by any deterioration in metabolic control, Ms. Hermann reported. The median baseline glycosylated hemoglobin (HbA1c) was 7.6% (59 mmol/mol) and this did not noticeably change at either the 6- or 12-month follow-up time point. Furthermore, the percentage of patients achieving a target HbA1c of less than 7.5% was 52% at baseline, 55% at 6 months, and 52% at 12 months.

The rate of diabetic ketoacidosis (DKA), defined as a pH of less than 7.3, also did not change significantly from baseline to the two follow-up points, with event rates of 1.5, 1.4, and 1.1, and the percentage of patients with at least one event being 0.8%, 0.5%, and 0.4%.

“We analyzed an overall well-controlled group of pediatric individuals with type 1 diabetes,” Ms. Hermann said. “We observed a significant reduction of severe hypoglycemia. We observed no improvements in HbA1c, but the overall HbA1c remained stable. We observed no statistically significant change in DKA.”

Future research will try to address some of the limitations that the current data may have, such as looking at a longer follow-up period and other endpoints such as hospitalization, Dr. Hermann suggested. Subgroup analyses are also planned.

“Large databases like the DPV registry ... provide a contemporary picture of diabetes treatment and how the use of diabetes technology has changed over the years and also how diabetes outcomes have improved over the years,” Ms. Hermann said. “It has yet to be seen how most recent changes and advances in diabetes technology will affect these long-term trends.”

The work was partially funded by the German Center for Diabetes Research, the German Diabetes Society, Abbott, and Sanofi. Dr. Hermann reported no personal conflicts of interest.
 

SOURCE: Hermann J et al. EASD 2018, Abstract 149.

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

 

BERLIN – A significant reduction in severe hypoglycemia was seen within the first year of using continuous glucose monitoring in a registry study of more than 3,000 children with type 1 diabetes mellitus.

Prior to continuous glucose monitoring (CGM) use, 3.9% of 3,171 individuals, aged a median of 12 years, had hypoglycemia events requiring external help. After 6 months of using CGM, however, the rate of severe hypoglycemia had fallen to 1.2% (P = .10), which remained at 1.2% at 12 months (P = .002). The event rate (events per 100 patient-years) was 10.6 at baseline, 7.8 at 6 months, and 6.1 at 12 months.

Fewer hypoglycemia events leading to coma or convulsion were seen with CGM over time, with 1.3%, 0.6%, and 0.7% of patients reporting at least one event at baseline, 6 months (P = .08), and 12 months (P = .15), respectively. Corresponding event rates were 2.5, 1.6, and 1.7 per 100 patient-years.

“The use of continuous glucose monitoring systems has increased considerably in the past years in individuals with type 1 diabetes,” noted Julia Hermann, a PhD student at Ulm University (Germany), at the annual meeting of the European Association for the Study of Diabetes. “In Germany, for example, CGM use rose considerably when reimbursement by health insurance for CGM started in the summer of 2016.”

Ms. Hermann noted that there were studies showing improved metabolic control with CGM but that clinical studies were often limited by the population of patients studied, restricting treatment to selected patients who may have been adhering better to the use of CGM because they were in a trial.

The aim of the present study was to assess metabolic control and acute complications associated with CGM use in children during its first year of use in a real-world population. Anonymized patient records from the prospective German-Austrian-Luxembourg diabetes patient follow-up (DPV) registry were used.

The DPV database is a standardized, computed-based registry of more than 500,000 adult and pediatric patients with all types of diabetes. It was established in 1995 and receives data from 471 diabetes clinics in Germany, Austria, Luxembourg, and Switzerland.

For the current analysis patients had to be aged under 18 years, have had type 1 diabetes mellitus for at least 1 year, have data available for the months prior to starting CGM, and have at least 1 year of follow-up. For inclusion there also had to be documented use of CMG for at least half of the observation time.

The reduction of severe hypoglycemia observed was not accompanied by any deterioration in metabolic control, Ms. Hermann reported. The median baseline glycosylated hemoglobin (HbA1c) was 7.6% (59 mmol/mol) and this did not noticeably change at either the 6- or 12-month follow-up time point. Furthermore, the percentage of patients achieving a target HbA1c of less than 7.5% was 52% at baseline, 55% at 6 months, and 52% at 12 months.

The rate of diabetic ketoacidosis (DKA), defined as a pH of less than 7.3, also did not change significantly from baseline to the two follow-up points, with event rates of 1.5, 1.4, and 1.1, and the percentage of patients with at least one event being 0.8%, 0.5%, and 0.4%.

“We analyzed an overall well-controlled group of pediatric individuals with type 1 diabetes,” Ms. Hermann said. “We observed a significant reduction of severe hypoglycemia. We observed no improvements in HbA1c, but the overall HbA1c remained stable. We observed no statistically significant change in DKA.”

Future research will try to address some of the limitations that the current data may have, such as looking at a longer follow-up period and other endpoints such as hospitalization, Dr. Hermann suggested. Subgroup analyses are also planned.

“Large databases like the DPV registry ... provide a contemporary picture of diabetes treatment and how the use of diabetes technology has changed over the years and also how diabetes outcomes have improved over the years,” Ms. Hermann said. “It has yet to be seen how most recent changes and advances in diabetes technology will affect these long-term trends.”

The work was partially funded by the German Center for Diabetes Research, the German Diabetes Society, Abbott, and Sanofi. Dr. Hermann reported no personal conflicts of interest.
 

SOURCE: Hermann J et al. EASD 2018, Abstract 149.

 

BERLIN – A significant reduction in severe hypoglycemia was seen within the first year of using continuous glucose monitoring in a registry study of more than 3,000 children with type 1 diabetes mellitus.

Prior to continuous glucose monitoring (CGM) use, 3.9% of 3,171 individuals, aged a median of 12 years, had hypoglycemia events requiring external help. After 6 months of using CGM, however, the rate of severe hypoglycemia had fallen to 1.2% (P = .10), which remained at 1.2% at 12 months (P = .002). The event rate (events per 100 patient-years) was 10.6 at baseline, 7.8 at 6 months, and 6.1 at 12 months.

Fewer hypoglycemia events leading to coma or convulsion were seen with CGM over time, with 1.3%, 0.6%, and 0.7% of patients reporting at least one event at baseline, 6 months (P = .08), and 12 months (P = .15), respectively. Corresponding event rates were 2.5, 1.6, and 1.7 per 100 patient-years.

“The use of continuous glucose monitoring systems has increased considerably in the past years in individuals with type 1 diabetes,” noted Julia Hermann, a PhD student at Ulm University (Germany), at the annual meeting of the European Association for the Study of Diabetes. “In Germany, for example, CGM use rose considerably when reimbursement by health insurance for CGM started in the summer of 2016.”

Ms. Hermann noted that there were studies showing improved metabolic control with CGM but that clinical studies were often limited by the population of patients studied, restricting treatment to selected patients who may have been adhering better to the use of CGM because they were in a trial.

The aim of the present study was to assess metabolic control and acute complications associated with CGM use in children during its first year of use in a real-world population. Anonymized patient records from the prospective German-Austrian-Luxembourg diabetes patient follow-up (DPV) registry were used.

The DPV database is a standardized, computed-based registry of more than 500,000 adult and pediatric patients with all types of diabetes. It was established in 1995 and receives data from 471 diabetes clinics in Germany, Austria, Luxembourg, and Switzerland.

For the current analysis patients had to be aged under 18 years, have had type 1 diabetes mellitus for at least 1 year, have data available for the months prior to starting CGM, and have at least 1 year of follow-up. For inclusion there also had to be documented use of CMG for at least half of the observation time.

The reduction of severe hypoglycemia observed was not accompanied by any deterioration in metabolic control, Ms. Hermann reported. The median baseline glycosylated hemoglobin (HbA1c) was 7.6% (59 mmol/mol) and this did not noticeably change at either the 6- or 12-month follow-up time point. Furthermore, the percentage of patients achieving a target HbA1c of less than 7.5% was 52% at baseline, 55% at 6 months, and 52% at 12 months.

The rate of diabetic ketoacidosis (DKA), defined as a pH of less than 7.3, also did not change significantly from baseline to the two follow-up points, with event rates of 1.5, 1.4, and 1.1, and the percentage of patients with at least one event being 0.8%, 0.5%, and 0.4%.

“We analyzed an overall well-controlled group of pediatric individuals with type 1 diabetes,” Ms. Hermann said. “We observed a significant reduction of severe hypoglycemia. We observed no improvements in HbA1c, but the overall HbA1c remained stable. We observed no statistically significant change in DKA.”

Future research will try to address some of the limitations that the current data may have, such as looking at a longer follow-up period and other endpoints such as hospitalization, Dr. Hermann suggested. Subgroup analyses are also planned.

“Large databases like the DPV registry ... provide a contemporary picture of diabetes treatment and how the use of diabetes technology has changed over the years and also how diabetes outcomes have improved over the years,” Ms. Hermann said. “It has yet to be seen how most recent changes and advances in diabetes technology will affect these long-term trends.”

The work was partially funded by the German Center for Diabetes Research, the German Diabetes Society, Abbott, and Sanofi. Dr. Hermann reported no personal conflicts of interest.
 

SOURCE: Hermann J et al. EASD 2018, Abstract 149.

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

REPORTING FROM EASD 2018

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
CME ID
189400
Vitals

 

Key clinical point: Continuous glucose monitoring helped to significantly reduce severe hypoglycemia episodes during the first year.

Major finding: A total of 3.9%, 1.2%, and 1.2% of patients experienced hypoglycemia needing external help at baseline, 6 months, and 12 months, respectively.

Study details: More than 3,000 pediatric patients using continuous glucose monitoring in the German-Austrian-Luxembourg diabetes patient follow-up registry.

Disclosures: The work was partially funded by the German Center for Diabetes Research, the German Diabetes Society, Abbott, and Sanofi. The presenting author reported no personal conflicts of interest.

Source: Hermann J et al. EASD 2018, Abstract 149.

Disqus Comments
Default
Use ProPublica

CDC: No medical therapy can yet be recommended for acute flaccid myelitis

Article Type
Changed
Fri, 01/18/2019 - 18:08

The updated guidance on managing acute flaccid myelitis is unlikely to relieve the frustrations of physicians struggling to treat the condition.

CDC News icon

After reviewing the extant data on the baffling disorder, the Centers for Disease Control and Prevention found no evidence that corticosteroids, interferon, antivirals, or any other immunologic or biologic therapy is an effective treatment.

All of the treatments mentioned in the guidance have been used anecdotally, and often for cases proven to be associated with enterovirus-related cases. However, there are no well validated studies confirming benefit for any of these approaches, the agency said in its clinical management document.

Acute flaccid myelitis (AFM) has stricken 90 patients in the United States this year and another 252 cases are being investigated, according to new data from the CDC. The number of confirmed cases is triple that seen in 2017. Whether the disease is an infectious or autoimmune process, or something else entirely, remains unknown.



In response to the outbreak – the largest since 2014 – an expert panel of 4 CDC staff physicians reviewed the literature to find what, if any, treatments were effective; another 14 external experts provided input on the recommendations. At this point, nothing can be officially recommended, the agency said.

Corticosteroids

Corticosteroids should not be administered to most patients with AFM. In addition to “a theoretical concern” about the potential adverse effects of these drugs in acute infections, there is some hard evidence that they are associated with worse outcomes in enteroviral neuroinvasive diseases, particularly those caused by EV-71.

This observation, following a 2012 outbreak in Cambodia, led a World Health Organization commission to conclude that corticosteroids were contraindicated in the management of EV-71–associated neuroinvasive disease. This year, there has been an uptick in EV-A71-associated neurologic disease.

The CDC did hedge its advice on corticosteroids a bit in the setting of AFM, however. “There may be theoretical benefit for steroids in the setting of severe cord swelling or long tract signs suggesting white matter involvement, where steroids may salvage tissue that may be harmed due to an ongoing immune/inflammatory response. While AFM is clinically and radiographically defined by the predominance of gray matter damage in the spinal cord, some patients may have some white matter involvement. It is not clear if these different patterns are important relative to therapeutic considerations.”

Nevertheless, the agency does not recommend corticosteroid use for these patients. “The possible benefits of the use of corticosteroids to manage spinal cord edema or white matter involvement in AFM should be balanced with the possible harm due to immunosuppression in the setting of possible viral infection.”
 

IVIG

While IVIG holds some theoretical benefit for AFM, there are no high-level human data, the guidelines state. The treatment is generally safe and well tolerated, but the few reports of its use in AFM did not show clear benefit. These include two case series. One suggested an acute improvement of neurologic status, but no long-term resolution of deficits. The other indicated neither significant improvement nor deterioration.

 

 

However, current practice at Children’s Hospital of Philadelphia is to initiate IVIG therapy at AFM diagnosis in hopes of boosting humoral immunity.

Nevertheless, the CDC said, “For IVIG to modify disease in an active viral infectious process, early administration is likely required, and possibly prior to exposure,” and the treatment cannot be recommended.
 

Plasma exchange

Plasma exchange in combination with IVIG and corticosteroids was ineffective in a case series of four Argentinian children, although a single case published last year found that the combination was associated with significant improvement. However, there are not enough data to recommend this approach.

Fluoxetine

Fluoxetine’s antiviral potential turned up in a high-throughput screening project to identify novel compounds with antiviral efficacy against enteroviruses. In 2012, researchers from the University of California, Los Angeles, tested more than 1,000 compounds and found that the SSRI is a potent inhibitor of coxsackievirus. A later project at the National Institutes of Health replicated this finding, and determined that fluoxetine inhibited several enteroviruses, including the AFM suspect, EV-D68.

Fluoxetine concentrates more highly in the central nervous system than it does in plasma, but its antiviral properties have nothing to do with neurotransmitter activity. Rather, it appears to inhibit protein 2C, a highly conserved nonstructural protein that’s crucial to the assembly of RNA into virion particles.

In early November, a retrospective study examined fluoxetine’s use in 30 AFM patients, compared with 26 who did not receive it. The primary outcome was change in summative limb strength score. The study did little to clarify any benefit, however. The authors concluded that fluoxetine was preferentially given to patients with EDV-68 infections. They had more severe impairment at nadir, and at the last follow-up of about 1 year, they had worse outcomes.

“There is no clear human evidence for efficacy of fluoxetine in the treatment of AFM based on a single retrospective evaluation conducted in patients with AFM, and data from a mouse model also did not support efficacy,” the CDC said.
 

Antiviral medications

The CDC is quite clear on its recommendation that these drugs are not indicated in AFM, since it is not yet proven to be an infectious process.

“Any guidance regarding antiviral medications should be interpreted with great caution, given the unknowns about the pathogenesis of this illness at present ... Testing has been conducted at CDC for antiviral activity of compounds pleconaril, pocapavir, and vapendavir and none have significant activity against currently circulating strains of EV-D68 at clinically relevant concentrations.”
 

Interferon

There is some anecdotal evidence that interferon alpha-2b was beneficial in treating a polio-like syndrome associated with West Nile virus and Saint Louis encephalitis. “Although there are limited in vitro, animal, and anecdotal human data suggesting activity of some interferons against viral infections, sufficient data are lacking in the setting of AFM,” the agency said. “There is no indication that interferon should be used for the treatment of AFM, and there is concern about the potential for harm from the use of interferon given the immunomodulatory effects in the setting of possible ongoing viral replication.”

SOURCE: CDC Acute Flaccid Myelitis: Interim Considerations for Clinical Management

Publications
Topics
Sections

The updated guidance on managing acute flaccid myelitis is unlikely to relieve the frustrations of physicians struggling to treat the condition.

CDC News icon

After reviewing the extant data on the baffling disorder, the Centers for Disease Control and Prevention found no evidence that corticosteroids, interferon, antivirals, or any other immunologic or biologic therapy is an effective treatment.

All of the treatments mentioned in the guidance have been used anecdotally, and often for cases proven to be associated with enterovirus-related cases. However, there are no well validated studies confirming benefit for any of these approaches, the agency said in its clinical management document.

Acute flaccid myelitis (AFM) has stricken 90 patients in the United States this year and another 252 cases are being investigated, according to new data from the CDC. The number of confirmed cases is triple that seen in 2017. Whether the disease is an infectious or autoimmune process, or something else entirely, remains unknown.



In response to the outbreak – the largest since 2014 – an expert panel of 4 CDC staff physicians reviewed the literature to find what, if any, treatments were effective; another 14 external experts provided input on the recommendations. At this point, nothing can be officially recommended, the agency said.

Corticosteroids

Corticosteroids should not be administered to most patients with AFM. In addition to “a theoretical concern” about the potential adverse effects of these drugs in acute infections, there is some hard evidence that they are associated with worse outcomes in enteroviral neuroinvasive diseases, particularly those caused by EV-71.

This observation, following a 2012 outbreak in Cambodia, led a World Health Organization commission to conclude that corticosteroids were contraindicated in the management of EV-71–associated neuroinvasive disease. This year, there has been an uptick in EV-A71-associated neurologic disease.

The CDC did hedge its advice on corticosteroids a bit in the setting of AFM, however. “There may be theoretical benefit for steroids in the setting of severe cord swelling or long tract signs suggesting white matter involvement, where steroids may salvage tissue that may be harmed due to an ongoing immune/inflammatory response. While AFM is clinically and radiographically defined by the predominance of gray matter damage in the spinal cord, some patients may have some white matter involvement. It is not clear if these different patterns are important relative to therapeutic considerations.”

Nevertheless, the agency does not recommend corticosteroid use for these patients. “The possible benefits of the use of corticosteroids to manage spinal cord edema or white matter involvement in AFM should be balanced with the possible harm due to immunosuppression in the setting of possible viral infection.”
 

IVIG

While IVIG holds some theoretical benefit for AFM, there are no high-level human data, the guidelines state. The treatment is generally safe and well tolerated, but the few reports of its use in AFM did not show clear benefit. These include two case series. One suggested an acute improvement of neurologic status, but no long-term resolution of deficits. The other indicated neither significant improvement nor deterioration.

 

 

However, current practice at Children’s Hospital of Philadelphia is to initiate IVIG therapy at AFM diagnosis in hopes of boosting humoral immunity.

Nevertheless, the CDC said, “For IVIG to modify disease in an active viral infectious process, early administration is likely required, and possibly prior to exposure,” and the treatment cannot be recommended.
 

Plasma exchange

Plasma exchange in combination with IVIG and corticosteroids was ineffective in a case series of four Argentinian children, although a single case published last year found that the combination was associated with significant improvement. However, there are not enough data to recommend this approach.

Fluoxetine

Fluoxetine’s antiviral potential turned up in a high-throughput screening project to identify novel compounds with antiviral efficacy against enteroviruses. In 2012, researchers from the University of California, Los Angeles, tested more than 1,000 compounds and found that the SSRI is a potent inhibitor of coxsackievirus. A later project at the National Institutes of Health replicated this finding, and determined that fluoxetine inhibited several enteroviruses, including the AFM suspect, EV-D68.

Fluoxetine concentrates more highly in the central nervous system than it does in plasma, but its antiviral properties have nothing to do with neurotransmitter activity. Rather, it appears to inhibit protein 2C, a highly conserved nonstructural protein that’s crucial to the assembly of RNA into virion particles.

In early November, a retrospective study examined fluoxetine’s use in 30 AFM patients, compared with 26 who did not receive it. The primary outcome was change in summative limb strength score. The study did little to clarify any benefit, however. The authors concluded that fluoxetine was preferentially given to patients with EDV-68 infections. They had more severe impairment at nadir, and at the last follow-up of about 1 year, they had worse outcomes.

“There is no clear human evidence for efficacy of fluoxetine in the treatment of AFM based on a single retrospective evaluation conducted in patients with AFM, and data from a mouse model also did not support efficacy,” the CDC said.
 

Antiviral medications

The CDC is quite clear on its recommendation that these drugs are not indicated in AFM, since it is not yet proven to be an infectious process.

“Any guidance regarding antiviral medications should be interpreted with great caution, given the unknowns about the pathogenesis of this illness at present ... Testing has been conducted at CDC for antiviral activity of compounds pleconaril, pocapavir, and vapendavir and none have significant activity against currently circulating strains of EV-D68 at clinically relevant concentrations.”
 

Interferon

There is some anecdotal evidence that interferon alpha-2b was beneficial in treating a polio-like syndrome associated with West Nile virus and Saint Louis encephalitis. “Although there are limited in vitro, animal, and anecdotal human data suggesting activity of some interferons against viral infections, sufficient data are lacking in the setting of AFM,” the agency said. “There is no indication that interferon should be used for the treatment of AFM, and there is concern about the potential for harm from the use of interferon given the immunomodulatory effects in the setting of possible ongoing viral replication.”

SOURCE: CDC Acute Flaccid Myelitis: Interim Considerations for Clinical Management

The updated guidance on managing acute flaccid myelitis is unlikely to relieve the frustrations of physicians struggling to treat the condition.

CDC News icon

After reviewing the extant data on the baffling disorder, the Centers for Disease Control and Prevention found no evidence that corticosteroids, interferon, antivirals, or any other immunologic or biologic therapy is an effective treatment.

All of the treatments mentioned in the guidance have been used anecdotally, and often for cases proven to be associated with enterovirus-related cases. However, there are no well validated studies confirming benefit for any of these approaches, the agency said in its clinical management document.

Acute flaccid myelitis (AFM) has stricken 90 patients in the United States this year and another 252 cases are being investigated, according to new data from the CDC. The number of confirmed cases is triple that seen in 2017. Whether the disease is an infectious or autoimmune process, or something else entirely, remains unknown.



In response to the outbreak – the largest since 2014 – an expert panel of 4 CDC staff physicians reviewed the literature to find what, if any, treatments were effective; another 14 external experts provided input on the recommendations. At this point, nothing can be officially recommended, the agency said.

Corticosteroids

Corticosteroids should not be administered to most patients with AFM. In addition to “a theoretical concern” about the potential adverse effects of these drugs in acute infections, there is some hard evidence that they are associated with worse outcomes in enteroviral neuroinvasive diseases, particularly those caused by EV-71.

This observation, following a 2012 outbreak in Cambodia, led a World Health Organization commission to conclude that corticosteroids were contraindicated in the management of EV-71–associated neuroinvasive disease. This year, there has been an uptick in EV-A71-associated neurologic disease.

The CDC did hedge its advice on corticosteroids a bit in the setting of AFM, however. “There may be theoretical benefit for steroids in the setting of severe cord swelling or long tract signs suggesting white matter involvement, where steroids may salvage tissue that may be harmed due to an ongoing immune/inflammatory response. While AFM is clinically and radiographically defined by the predominance of gray matter damage in the spinal cord, some patients may have some white matter involvement. It is not clear if these different patterns are important relative to therapeutic considerations.”

Nevertheless, the agency does not recommend corticosteroid use for these patients. “The possible benefits of the use of corticosteroids to manage spinal cord edema or white matter involvement in AFM should be balanced with the possible harm due to immunosuppression in the setting of possible viral infection.”
 

IVIG

While IVIG holds some theoretical benefit for AFM, there are no high-level human data, the guidelines state. The treatment is generally safe and well tolerated, but the few reports of its use in AFM did not show clear benefit. These include two case series. One suggested an acute improvement of neurologic status, but no long-term resolution of deficits. The other indicated neither significant improvement nor deterioration.

 

 

However, current practice at Children’s Hospital of Philadelphia is to initiate IVIG therapy at AFM diagnosis in hopes of boosting humoral immunity.

Nevertheless, the CDC said, “For IVIG to modify disease in an active viral infectious process, early administration is likely required, and possibly prior to exposure,” and the treatment cannot be recommended.
 

Plasma exchange

Plasma exchange in combination with IVIG and corticosteroids was ineffective in a case series of four Argentinian children, although a single case published last year found that the combination was associated with significant improvement. However, there are not enough data to recommend this approach.

Fluoxetine

Fluoxetine’s antiviral potential turned up in a high-throughput screening project to identify novel compounds with antiviral efficacy against enteroviruses. In 2012, researchers from the University of California, Los Angeles, tested more than 1,000 compounds and found that the SSRI is a potent inhibitor of coxsackievirus. A later project at the National Institutes of Health replicated this finding, and determined that fluoxetine inhibited several enteroviruses, including the AFM suspect, EV-D68.

Fluoxetine concentrates more highly in the central nervous system than it does in plasma, but its antiviral properties have nothing to do with neurotransmitter activity. Rather, it appears to inhibit protein 2C, a highly conserved nonstructural protein that’s crucial to the assembly of RNA into virion particles.

In early November, a retrospective study examined fluoxetine’s use in 30 AFM patients, compared with 26 who did not receive it. The primary outcome was change in summative limb strength score. The study did little to clarify any benefit, however. The authors concluded that fluoxetine was preferentially given to patients with EDV-68 infections. They had more severe impairment at nadir, and at the last follow-up of about 1 year, they had worse outcomes.

“There is no clear human evidence for efficacy of fluoxetine in the treatment of AFM based on a single retrospective evaluation conducted in patients with AFM, and data from a mouse model also did not support efficacy,” the CDC said.
 

Antiviral medications

The CDC is quite clear on its recommendation that these drugs are not indicated in AFM, since it is not yet proven to be an infectious process.

“Any guidance regarding antiviral medications should be interpreted with great caution, given the unknowns about the pathogenesis of this illness at present ... Testing has been conducted at CDC for antiviral activity of compounds pleconaril, pocapavir, and vapendavir and none have significant activity against currently circulating strains of EV-D68 at clinically relevant concentrations.”
 

Interferon

There is some anecdotal evidence that interferon alpha-2b was beneficial in treating a polio-like syndrome associated with West Nile virus and Saint Louis encephalitis. “Although there are limited in vitro, animal, and anecdotal human data suggesting activity of some interferons against viral infections, sufficient data are lacking in the setting of AFM,” the agency said. “There is no indication that interferon should be used for the treatment of AFM, and there is concern about the potential for harm from the use of interferon given the immunomodulatory effects in the setting of possible ongoing viral replication.”

SOURCE: CDC Acute Flaccid Myelitis: Interim Considerations for Clinical Management

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

Probiotics and pediatric gastroenteritis

Article Type
Changed
Wed, 05/26/2021 - 13:48

 

Two probiotic products did not significantly impact children with gastroenteritis. Also today, all patients with VTE have a high risk of recurrence, the REDUCE-IT study establishes a “phenomenal” triglyceride theory, and new data on patients with gout suggest a treat-to-target approach.

Amazon Alexa
Apple Podcasts

Google Podcasts
Spotify

Publications
Topics
Sections

 

Two probiotic products did not significantly impact children with gastroenteritis. Also today, all patients with VTE have a high risk of recurrence, the REDUCE-IT study establishes a “phenomenal” triglyceride theory, and new data on patients with gout suggest a treat-to-target approach.

Amazon Alexa
Apple Podcasts

Google Podcasts
Spotify

 

Two probiotic products did not significantly impact children with gastroenteritis. Also today, all patients with VTE have a high risk of recurrence, the REDUCE-IT study establishes a “phenomenal” triglyceride theory, and new data on patients with gout suggest a treat-to-target approach.

Amazon Alexa
Apple Podcasts

Google Podcasts
Spotify

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

Cancer Survivors’ Risk of Mood Disorders

Article Type
Changed
Thu, 12/15/2022 - 14:43
Researchers examine the risk of mood disorders in patients who survived cancer years after diagnosis.

Cancer survivors have a higher risk of depression within 2 years after the diagnosis, according to a meta-analysis. But is that true of survivors of all types of cancer? In fact, risk is multifactorial because patients, cancers, comorbidities, and impacts of treatments are all different, say researchers who conducted a study to compare the risk of mood disorders longitudinally.

They matched 190,748 survivors with controls from the Taiwan National Health Insurance Research Database. The median follow-up times were 8.13 and 8.49 years, respectively. The 3 most common cancers were breast, colorectal, and head and neck. Surgery alone was the main treatment, followed by combinations of surgery, chemotherapy, and radiation.

Survivors had a significantly higher risk of mood disorders: 8.38 per 1,000 person-years, compared with 7.21 in the control patients. Major depression and depression disorder were the most common subtypes.

However, the risk of mood disorders (1.13-fold) peaked during the year after the index date and declined thereafter. Moreover, 2 and 5 years later, the risk was similar between the 2 groups. And after 5 years, the risk was even lower in the survivor group than in the control group.

The researchers found patients fell into 3 main categories: persistently increasing risk, higher risk in the first few years and after 5 years of follow-up, and higher risk in the first few years but no difference thereafter. Patients with head and neck cancer, nasopharyngeal cancer, and esophageal cancer were in the first group, with distinct longitudinal patterns. Their risk at 5 years was greater than that of the general population.

Being female, aged 40-59 years, having > 2 primary cancers, having ≥ 2 treatment modalities, Charlson comorbidity index scores > 3, higher urbanization level, and lower income levels were independent risk factors for mood disorders.

The researchers say their findings highlight the importance of taking follow-up time, cancer types, and cancer-related treatment into consideration when evaluating mood disorders in cancer survivors. They also emphasize the need for better psychological management not only in the early postdiagnosis years, but in late follow-up for patients with a “persistent” risk.

Source:
Huang WK, Juang YY, Chung CC, et al. J Affect Disord. 2018;236:80-87.

Publications
Topics
Sections
Researchers examine the risk of mood disorders in patients who survived cancer years after diagnosis.
Researchers examine the risk of mood disorders in patients who survived cancer years after diagnosis.

Cancer survivors have a higher risk of depression within 2 years after the diagnosis, according to a meta-analysis. But is that true of survivors of all types of cancer? In fact, risk is multifactorial because patients, cancers, comorbidities, and impacts of treatments are all different, say researchers who conducted a study to compare the risk of mood disorders longitudinally.

They matched 190,748 survivors with controls from the Taiwan National Health Insurance Research Database. The median follow-up times were 8.13 and 8.49 years, respectively. The 3 most common cancers were breast, colorectal, and head and neck. Surgery alone was the main treatment, followed by combinations of surgery, chemotherapy, and radiation.

Survivors had a significantly higher risk of mood disorders: 8.38 per 1,000 person-years, compared with 7.21 in the control patients. Major depression and depression disorder were the most common subtypes.

However, the risk of mood disorders (1.13-fold) peaked during the year after the index date and declined thereafter. Moreover, 2 and 5 years later, the risk was similar between the 2 groups. And after 5 years, the risk was even lower in the survivor group than in the control group.

The researchers found patients fell into 3 main categories: persistently increasing risk, higher risk in the first few years and after 5 years of follow-up, and higher risk in the first few years but no difference thereafter. Patients with head and neck cancer, nasopharyngeal cancer, and esophageal cancer were in the first group, with distinct longitudinal patterns. Their risk at 5 years was greater than that of the general population.

Being female, aged 40-59 years, having > 2 primary cancers, having ≥ 2 treatment modalities, Charlson comorbidity index scores > 3, higher urbanization level, and lower income levels were independent risk factors for mood disorders.

The researchers say their findings highlight the importance of taking follow-up time, cancer types, and cancer-related treatment into consideration when evaluating mood disorders in cancer survivors. They also emphasize the need for better psychological management not only in the early postdiagnosis years, but in late follow-up for patients with a “persistent” risk.

Source:
Huang WK, Juang YY, Chung CC, et al. J Affect Disord. 2018;236:80-87.

Cancer survivors have a higher risk of depression within 2 years after the diagnosis, according to a meta-analysis. But is that true of survivors of all types of cancer? In fact, risk is multifactorial because patients, cancers, comorbidities, and impacts of treatments are all different, say researchers who conducted a study to compare the risk of mood disorders longitudinally.

They matched 190,748 survivors with controls from the Taiwan National Health Insurance Research Database. The median follow-up times were 8.13 and 8.49 years, respectively. The 3 most common cancers were breast, colorectal, and head and neck. Surgery alone was the main treatment, followed by combinations of surgery, chemotherapy, and radiation.

Survivors had a significantly higher risk of mood disorders: 8.38 per 1,000 person-years, compared with 7.21 in the control patients. Major depression and depression disorder were the most common subtypes.

However, the risk of mood disorders (1.13-fold) peaked during the year after the index date and declined thereafter. Moreover, 2 and 5 years later, the risk was similar between the 2 groups. And after 5 years, the risk was even lower in the survivor group than in the control group.

The researchers found patients fell into 3 main categories: persistently increasing risk, higher risk in the first few years and after 5 years of follow-up, and higher risk in the first few years but no difference thereafter. Patients with head and neck cancer, nasopharyngeal cancer, and esophageal cancer were in the first group, with distinct longitudinal patterns. Their risk at 5 years was greater than that of the general population.

Being female, aged 40-59 years, having > 2 primary cancers, having ≥ 2 treatment modalities, Charlson comorbidity index scores > 3, higher urbanization level, and lower income levels were independent risk factors for mood disorders.

The researchers say their findings highlight the importance of taking follow-up time, cancer types, and cancer-related treatment into consideration when evaluating mood disorders in cancer survivors. They also emphasize the need for better psychological management not only in the early postdiagnosis years, but in late follow-up for patients with a “persistent” risk.

Source:
Huang WK, Juang YY, Chung CC, et al. J Affect Disord. 2018;236:80-87.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Mon, 11/19/2018 - 14:30
Un-Gate On Date
Mon, 11/19/2018 - 14:30
Use ProPublica
CFC Schedule Remove Status
Mon, 11/19/2018 - 14:30