User login
What’s Eating You? Oriental Rat Flea (Xenopsylla cheopis)
A dult Siphonaptera (fleas) are highly adapted to life on the surface of their hosts. Their small 2- to 10-mm bodies are laterally flattened and wingless. They utilize particularly strong hind legs for jumping up to 150 times their body length and backward-directed spines on their legs and bodies for moving forward through fur, hair, and feathers. Xenopsylla cheopis , the oriental rat flea, lacks pronotal and genal combs and has a mesopleuron divided by internal scleritinization (Figure). These features differentiate the species from its close relatives, Ctenocephalides (cat and dog fleas), which have both sets of combs, as well as Pulex irritans (human flea), which do not have a divided mesopleuron. 1,2
Flea-borne infections are extremely important to public health and are present throughout the world. Further, humidity and warmth are essential for the life cycle of many species of fleas. Predicted global warming likely will increase their distribution, allowing the spread of diseases they carry into previously untouched areas.1 Therefore, it is important to continue to examine species that carry particularly dangerous pathogens, such as X cheopis.
Disease Vector
Xenopsylla cheopis primarily is known for being a vector in the transmission of Yersinia pestis, the etiologic agent of the plague. Plague occurs in 3 forms: bubonic, pneumonic, and septicemic. It has caused major epidemics throughout history, the most widely known being the Black Death, which lasted for 130 years, beginning in the 1330s in China and spreading into Europe where it wiped out one-third of the population. However, bubonic plague is thought to have caused documented outbreaks as early as 320
Between January 2010 and December 2015, 3248 cases of plague in humans were reported, resulting in 584 deaths worldwide.5 It is thought that the plague originated in Central Asia, and this area still is a focus of disease. However, the at-risk population is reduced to breeders and hunters of gerbils and marmots, the main reservoirs in the area. In Africa, 4 countries still regularly report cases, with Madagascar being the most severely affected country in the world.5 The Democratic Republic of the Congo, Uganda, and Tanzania also are affected. The Americas also experience the plague. There are sporadic cases of bubonic plague in the northwest corner of Peru, mostly in rural areas. In the western United States, plague circulates among wild rodents, resulting in several reported cases each year, with the most recent confirmed case noted in California in August 2020.5,6 Further adding to its relevance, Y pestis is one of several organisms most likely to be used as a biologic weapon.3,4
Due to the historical and continued significance of Y pestis, many studies have been performed over the decades regarding its association with X cheopis. It has been discovered that fleas transmit the bacteria to the host in 2 ways. The most well-defined form of transmission occurs after an incubation period of Y pestis in the flea for 7 to 31 days. During this time, the bacteria form a dense biofilm on a valve in the flea foregut—the proventriculus—interfering with its function, which allows regurgitation of the blood and the bacteria it contains into the bite site and consequently disease transmission. The proventriculus can become completely blocked in some fleas, preventing any blood from reaching the midgut and causing starvation. In these scenarios, the flea will make continuous attempts to feed, increasing transmission.7 The hemin storage gene, hms, encoding the second messenger molecule cyclic di-GMP plays a critical role in biofilm formation and proventricular blockage.8 The phosphoheptose isomerase gene, GmhA, also has been elucidated as crucial in late transmission due to its role in biofilm formation.9 Early-phase transmission, or biofilm-independent transmission, has been documented to occur as early as 3 hours after infection of the flea but can occur for up to 4 days.10 Historically, the importance of early-phase transmission has been overlooked. Research has shown that it likely is crucial to the epizootic transmission of the plague.10 As a result, the search has begun for genes that contribute to the maintenance of Y pestis in the flea vector during the first 4 days of colonization. It is thought that a key evolutionary development was the selective loss-of-function mutation in a gene essential for the activity of urease, an enzyme that causes acute oral toxicity and mortality in fleas.11,12 The Yersinia murine toxin gene, Ymt, also allows for early survival of Y pestis in the flea midgut by producing a phospholipase D that protects the bacteria from toxic by-products produced during digestion of blood.11,13 In addition, gene products that function in lipid A modification are crucial for the ability of Y pestis to resist the action of cationic antimicrobial peptides it produces, such as cecropin A and polymyxin B.13
Murine typhus, an acute febrile illness caused by Rickettsia typhi, is another disease that can be spread by oriental rat fleas. It has a worldwide distribution. In the United States, R typhi–induced rickettsia mainly is concentrated in suburban areas of Texas and California where it is thought to be mainly spread by Ctenocephalides, but it also is found in Hawaii where spread by X cheopis has been documented.14,15 The most common symptoms of rickettsia include fever, headache, arthralgia, and a characteristic rash that is pruritic and maculopapular, starting on the trunk and spreading peripherally but sparing the palms and soles. This rash occurs about a week after the onset of fever.14Rickettsia felis also has been isolated in the oriental rat flea. However, only a handful of cases of human disease caused by this bacterium have been reported throughout the world, with clinical similarity to murine typhus likely leading to underestimation of disease prevalence.15Bartonella and other species of bacteria also have been documented to be spread by X cheopis.16 Unfortunately, the interactions of X cheopis with these other bacteria are not as well studied as its relationship with Y pestis.
Adverse Reactions
A flea bite itself can cause discomfort. It begins as a punctate hemorrhagic area that develops a surrounding wheal within 30 minutes. Over the course of 1 to 2 days, a delayed reaction occurs and there is a transition to an extremely pruritic, papular lesion. Bites often occur in clusters and can persist for weeks.1
Prevention and Treatment
Control of host animals via extermination and proper sanitation can secondarily reduce the population of X cheopis. Direct pesticide control of the flea population also has been suggested to reduce flea-borne disease. However, insecticides cause a selective pressure on the flea population, leading to populations that are resistant to them. For example, the flea population in Madagascar developed resistance to DDT (dichlorodiphenyltrichloroethane), dieldrin, deltamethrin, and cyfluthrin after their widespread use.17 Further, a recent study revealed resistance of X cheopis populations to alphacypermethrin, lambda-cyhalothrin, and etofenprox, none of which were used in mass vector control, indicating that some cross-resistance mechanism between these and the extensively used insecticides may exist. With the development of widespread resistance to most pesticides, flea control in endemic areas is difficult. Insecticide targeting to fleas on rodents (eg, rodent bait containing insecticide) can allow for more targeted insecticide treatment, limiting the development of resistance.17 Recent development of a maceration protocol used to detect zoonotic pathogens in fleas in the field also will allow management with pesticides to be targeted geographically and temporally where infected vectors are located.18 Research of the interaction between vector, pathogen, and insect microbiome also should continue, as it may allow for development of biopesticides, limiting the use of chemical pesticides all together. The strategy is based on the introduction of microorganisms that can reduce vector lifespan or their ability to transmit pathogens.17
When flea-transmitted diseases do occur, treatment with antibiotics is advised. Early treatment of the plague with effective antibiotics such as streptomycin, gentamicin, tetracycline, or chloramphenicol for a minimum of 10 days is critical for survival. Additionally, patients with bubonic plague should be isolated for at least 2 days after administration of antibiotics, while patients with the pneumonic form should be isolated for 4 days into therapy to prevent the spread of disease. Prophylactic therapy for individuals who come into contact with infected individuals also is advised.4 Patients with murine typhus typically respond to doxycycline, tetracycline, or fluoroquinolones. The few cases of R felis–induced disease have responded to doxycycline. Of note, short courses of treatment of doxycycline are appropriate and safe in young children. The short (3–7 day) nature of the course limits the chances of teeth staining.14
- Bitam I, Dittmar K, Parola P, et al. Flea and flea-borne diseases. Int J Infect Dis. 2010;14:E667-E676.
- Mathison BA, Pritt BS. Laboratory identification of arthropod ectoparasites. Clin Microbiol Rev. 2014;27:48-67.
- Ligon BL. Plague: a review of its history and potential as a biological weapon. Semin Pediatr Infect Dis. 2006;17:161-170.
- Josko D. Yersinia pestis: still a plague in the 21st century. Clin Lab Sci. 2004;17:25-29.
- Plague around the world, 2010–2015. Wkly Epidemiol Rec. 2016;91:89-93.
- Sullivan K. California confirms first human case of the plague in 5 years: what to know. NBC News website. https://www.nbcnews.com/health/health-news/california-confirms-first-human-case-bubonic-plague-5-years-what-n1237306. Published August 19, 2020. Accessed August 24, 2020.
- Hinnebusch BJ, Bland DM, Bosio CF, et al. Comparative ability of Oropsylla and Xenopsylla cheopis fleas to transmit Yersinia pestis by two different mechanisms. PLOS Negl Trop Dis. 2017;11:e0005276.
- Bobrov AG, Kirillina O, Vadyvaloo V, et al. The Yersinia pestis HmsCDE regulatory system is essential for blockage of the oriental rat flea (Xenopsylla cheopis), a classic plague vector. Environ Microbiol. 2015;17:947-959.
- Darby C, Ananth SL, Tan L, et al. Identification of gmhA, a Yersina pestis gene required for flea blockage, by using a Caenorhabditis elegans biofilm system. Infect Immun. 2005;73:7236-7242.
- Eisen RJ, Dennis DT, Gage KL. The role of early-phase transmission in the spread of Yersinia pestis. J Med Entomol. 2015;52:1183-1192.
- Carniel E. Subtle genetic modifications transformed an enteropathogen into a flea-borne pathogen. Proc Natl Acad Sci U S A. 2014;111:18409-18410.
- Chouikha I, Hinnebusch BJ. Silencing urease: a key evolutionary step that facilitated the adaptation of Yersinia pestis to the flea-borne transmission route. Proc Natl Acad Sci U S A. 2014;111:18709-19714.
- Aoyagi KL, Brooks BD, Bearden SW, et al. LPS modification promotes maintenance of Yersinia pestis in fleas. Microbiology. 2015;161:628-638.
- Civen R, Ngo V. Murine typhus: an unrecognized suburban vectorborne disease. Clin Infect Dis. 2008;46:913-918.
- Eremeeva ME, Warashina WR, Sturgeon MM, et al. Rickettsia typhi and R. felis in rat fleas (Xenopsylla cheopis), Oahu, Hawaii. Emerg Infect Dis. 2018;14:1613-1615.
- Billeter SA, Gundi VAKB, Rood MP, et al. Molecular detection and identification of Bartonella species in Xenopsylla cheopis fleas (Siphonaptera: Pulicidae) collected from Rattus norvecus rats in Los Angeles, California. Appl Environ Microbiol. 2011;77:7850-7852.
- Miarinjara A, Boyer S. Current perspectives on plague vector control in Madagascar: susceptibility status of Xenopsylla cheopis to 12 insecticides. PLOS Negl Trop Dis. 2016;10:e0004414.
- Harrison GF, Scheirer JL, Melanson VR. Development and validation of an arthropod maceration protocol for zoonotic pathogen detection in mosquitoes and fleas. J Vector Ecol. 2014;40:83-89.
A dult Siphonaptera (fleas) are highly adapted to life on the surface of their hosts. Their small 2- to 10-mm bodies are laterally flattened and wingless. They utilize particularly strong hind legs for jumping up to 150 times their body length and backward-directed spines on their legs and bodies for moving forward through fur, hair, and feathers. Xenopsylla cheopis , the oriental rat flea, lacks pronotal and genal combs and has a mesopleuron divided by internal scleritinization (Figure). These features differentiate the species from its close relatives, Ctenocephalides (cat and dog fleas), which have both sets of combs, as well as Pulex irritans (human flea), which do not have a divided mesopleuron. 1,2
Flea-borne infections are extremely important to public health and are present throughout the world. Further, humidity and warmth are essential for the life cycle of many species of fleas. Predicted global warming likely will increase their distribution, allowing the spread of diseases they carry into previously untouched areas.1 Therefore, it is important to continue to examine species that carry particularly dangerous pathogens, such as X cheopis.
Disease Vector
Xenopsylla cheopis primarily is known for being a vector in the transmission of Yersinia pestis, the etiologic agent of the plague. Plague occurs in 3 forms: bubonic, pneumonic, and septicemic. It has caused major epidemics throughout history, the most widely known being the Black Death, which lasted for 130 years, beginning in the 1330s in China and spreading into Europe where it wiped out one-third of the population. However, bubonic plague is thought to have caused documented outbreaks as early as 320
Between January 2010 and December 2015, 3248 cases of plague in humans were reported, resulting in 584 deaths worldwide.5 It is thought that the plague originated in Central Asia, and this area still is a focus of disease. However, the at-risk population is reduced to breeders and hunters of gerbils and marmots, the main reservoirs in the area. In Africa, 4 countries still regularly report cases, with Madagascar being the most severely affected country in the world.5 The Democratic Republic of the Congo, Uganda, and Tanzania also are affected. The Americas also experience the plague. There are sporadic cases of bubonic plague in the northwest corner of Peru, mostly in rural areas. In the western United States, plague circulates among wild rodents, resulting in several reported cases each year, with the most recent confirmed case noted in California in August 2020.5,6 Further adding to its relevance, Y pestis is one of several organisms most likely to be used as a biologic weapon.3,4
Due to the historical and continued significance of Y pestis, many studies have been performed over the decades regarding its association with X cheopis. It has been discovered that fleas transmit the bacteria to the host in 2 ways. The most well-defined form of transmission occurs after an incubation period of Y pestis in the flea for 7 to 31 days. During this time, the bacteria form a dense biofilm on a valve in the flea foregut—the proventriculus—interfering with its function, which allows regurgitation of the blood and the bacteria it contains into the bite site and consequently disease transmission. The proventriculus can become completely blocked in some fleas, preventing any blood from reaching the midgut and causing starvation. In these scenarios, the flea will make continuous attempts to feed, increasing transmission.7 The hemin storage gene, hms, encoding the second messenger molecule cyclic di-GMP plays a critical role in biofilm formation and proventricular blockage.8 The phosphoheptose isomerase gene, GmhA, also has been elucidated as crucial in late transmission due to its role in biofilm formation.9 Early-phase transmission, or biofilm-independent transmission, has been documented to occur as early as 3 hours after infection of the flea but can occur for up to 4 days.10 Historically, the importance of early-phase transmission has been overlooked. Research has shown that it likely is crucial to the epizootic transmission of the plague.10 As a result, the search has begun for genes that contribute to the maintenance of Y pestis in the flea vector during the first 4 days of colonization. It is thought that a key evolutionary development was the selective loss-of-function mutation in a gene essential for the activity of urease, an enzyme that causes acute oral toxicity and mortality in fleas.11,12 The Yersinia murine toxin gene, Ymt, also allows for early survival of Y pestis in the flea midgut by producing a phospholipase D that protects the bacteria from toxic by-products produced during digestion of blood.11,13 In addition, gene products that function in lipid A modification are crucial for the ability of Y pestis to resist the action of cationic antimicrobial peptides it produces, such as cecropin A and polymyxin B.13
Murine typhus, an acute febrile illness caused by Rickettsia typhi, is another disease that can be spread by oriental rat fleas. It has a worldwide distribution. In the United States, R typhi–induced rickettsia mainly is concentrated in suburban areas of Texas and California where it is thought to be mainly spread by Ctenocephalides, but it also is found in Hawaii where spread by X cheopis has been documented.14,15 The most common symptoms of rickettsia include fever, headache, arthralgia, and a characteristic rash that is pruritic and maculopapular, starting on the trunk and spreading peripherally but sparing the palms and soles. This rash occurs about a week after the onset of fever.14Rickettsia felis also has been isolated in the oriental rat flea. However, only a handful of cases of human disease caused by this bacterium have been reported throughout the world, with clinical similarity to murine typhus likely leading to underestimation of disease prevalence.15Bartonella and other species of bacteria also have been documented to be spread by X cheopis.16 Unfortunately, the interactions of X cheopis with these other bacteria are not as well studied as its relationship with Y pestis.
Adverse Reactions
A flea bite itself can cause discomfort. It begins as a punctate hemorrhagic area that develops a surrounding wheal within 30 minutes. Over the course of 1 to 2 days, a delayed reaction occurs and there is a transition to an extremely pruritic, papular lesion. Bites often occur in clusters and can persist for weeks.1
Prevention and Treatment
Control of host animals via extermination and proper sanitation can secondarily reduce the population of X cheopis. Direct pesticide control of the flea population also has been suggested to reduce flea-borne disease. However, insecticides cause a selective pressure on the flea population, leading to populations that are resistant to them. For example, the flea population in Madagascar developed resistance to DDT (dichlorodiphenyltrichloroethane), dieldrin, deltamethrin, and cyfluthrin after their widespread use.17 Further, a recent study revealed resistance of X cheopis populations to alphacypermethrin, lambda-cyhalothrin, and etofenprox, none of which were used in mass vector control, indicating that some cross-resistance mechanism between these and the extensively used insecticides may exist. With the development of widespread resistance to most pesticides, flea control in endemic areas is difficult. Insecticide targeting to fleas on rodents (eg, rodent bait containing insecticide) can allow for more targeted insecticide treatment, limiting the development of resistance.17 Recent development of a maceration protocol used to detect zoonotic pathogens in fleas in the field also will allow management with pesticides to be targeted geographically and temporally where infected vectors are located.18 Research of the interaction between vector, pathogen, and insect microbiome also should continue, as it may allow for development of biopesticides, limiting the use of chemical pesticides all together. The strategy is based on the introduction of microorganisms that can reduce vector lifespan or their ability to transmit pathogens.17
When flea-transmitted diseases do occur, treatment with antibiotics is advised. Early treatment of the plague with effective antibiotics such as streptomycin, gentamicin, tetracycline, or chloramphenicol for a minimum of 10 days is critical for survival. Additionally, patients with bubonic plague should be isolated for at least 2 days after administration of antibiotics, while patients with the pneumonic form should be isolated for 4 days into therapy to prevent the spread of disease. Prophylactic therapy for individuals who come into contact with infected individuals also is advised.4 Patients with murine typhus typically respond to doxycycline, tetracycline, or fluoroquinolones. The few cases of R felis–induced disease have responded to doxycycline. Of note, short courses of treatment of doxycycline are appropriate and safe in young children. The short (3–7 day) nature of the course limits the chances of teeth staining.14
A dult Siphonaptera (fleas) are highly adapted to life on the surface of their hosts. Their small 2- to 10-mm bodies are laterally flattened and wingless. They utilize particularly strong hind legs for jumping up to 150 times their body length and backward-directed spines on their legs and bodies for moving forward through fur, hair, and feathers. Xenopsylla cheopis , the oriental rat flea, lacks pronotal and genal combs and has a mesopleuron divided by internal scleritinization (Figure). These features differentiate the species from its close relatives, Ctenocephalides (cat and dog fleas), which have both sets of combs, as well as Pulex irritans (human flea), which do not have a divided mesopleuron. 1,2
Flea-borne infections are extremely important to public health and are present throughout the world. Further, humidity and warmth are essential for the life cycle of many species of fleas. Predicted global warming likely will increase their distribution, allowing the spread of diseases they carry into previously untouched areas.1 Therefore, it is important to continue to examine species that carry particularly dangerous pathogens, such as X cheopis.
Disease Vector
Xenopsylla cheopis primarily is known for being a vector in the transmission of Yersinia pestis, the etiologic agent of the plague. Plague occurs in 3 forms: bubonic, pneumonic, and septicemic. It has caused major epidemics throughout history, the most widely known being the Black Death, which lasted for 130 years, beginning in the 1330s in China and spreading into Europe where it wiped out one-third of the population. However, bubonic plague is thought to have caused documented outbreaks as early as 320
Between January 2010 and December 2015, 3248 cases of plague in humans were reported, resulting in 584 deaths worldwide.5 It is thought that the plague originated in Central Asia, and this area still is a focus of disease. However, the at-risk population is reduced to breeders and hunters of gerbils and marmots, the main reservoirs in the area. In Africa, 4 countries still regularly report cases, with Madagascar being the most severely affected country in the world.5 The Democratic Republic of the Congo, Uganda, and Tanzania also are affected. The Americas also experience the plague. There are sporadic cases of bubonic plague in the northwest corner of Peru, mostly in rural areas. In the western United States, plague circulates among wild rodents, resulting in several reported cases each year, with the most recent confirmed case noted in California in August 2020.5,6 Further adding to its relevance, Y pestis is one of several organisms most likely to be used as a biologic weapon.3,4
Due to the historical and continued significance of Y pestis, many studies have been performed over the decades regarding its association with X cheopis. It has been discovered that fleas transmit the bacteria to the host in 2 ways. The most well-defined form of transmission occurs after an incubation period of Y pestis in the flea for 7 to 31 days. During this time, the bacteria form a dense biofilm on a valve in the flea foregut—the proventriculus—interfering with its function, which allows regurgitation of the blood and the bacteria it contains into the bite site and consequently disease transmission. The proventriculus can become completely blocked in some fleas, preventing any blood from reaching the midgut and causing starvation. In these scenarios, the flea will make continuous attempts to feed, increasing transmission.7 The hemin storage gene, hms, encoding the second messenger molecule cyclic di-GMP plays a critical role in biofilm formation and proventricular blockage.8 The phosphoheptose isomerase gene, GmhA, also has been elucidated as crucial in late transmission due to its role in biofilm formation.9 Early-phase transmission, or biofilm-independent transmission, has been documented to occur as early as 3 hours after infection of the flea but can occur for up to 4 days.10 Historically, the importance of early-phase transmission has been overlooked. Research has shown that it likely is crucial to the epizootic transmission of the plague.10 As a result, the search has begun for genes that contribute to the maintenance of Y pestis in the flea vector during the first 4 days of colonization. It is thought that a key evolutionary development was the selective loss-of-function mutation in a gene essential for the activity of urease, an enzyme that causes acute oral toxicity and mortality in fleas.11,12 The Yersinia murine toxin gene, Ymt, also allows for early survival of Y pestis in the flea midgut by producing a phospholipase D that protects the bacteria from toxic by-products produced during digestion of blood.11,13 In addition, gene products that function in lipid A modification are crucial for the ability of Y pestis to resist the action of cationic antimicrobial peptides it produces, such as cecropin A and polymyxin B.13
Murine typhus, an acute febrile illness caused by Rickettsia typhi, is another disease that can be spread by oriental rat fleas. It has a worldwide distribution. In the United States, R typhi–induced rickettsia mainly is concentrated in suburban areas of Texas and California where it is thought to be mainly spread by Ctenocephalides, but it also is found in Hawaii where spread by X cheopis has been documented.14,15 The most common symptoms of rickettsia include fever, headache, arthralgia, and a characteristic rash that is pruritic and maculopapular, starting on the trunk and spreading peripherally but sparing the palms and soles. This rash occurs about a week after the onset of fever.14Rickettsia felis also has been isolated in the oriental rat flea. However, only a handful of cases of human disease caused by this bacterium have been reported throughout the world, with clinical similarity to murine typhus likely leading to underestimation of disease prevalence.15Bartonella and other species of bacteria also have been documented to be spread by X cheopis.16 Unfortunately, the interactions of X cheopis with these other bacteria are not as well studied as its relationship with Y pestis.
Adverse Reactions
A flea bite itself can cause discomfort. It begins as a punctate hemorrhagic area that develops a surrounding wheal within 30 minutes. Over the course of 1 to 2 days, a delayed reaction occurs and there is a transition to an extremely pruritic, papular lesion. Bites often occur in clusters and can persist for weeks.1
Prevention and Treatment
Control of host animals via extermination and proper sanitation can secondarily reduce the population of X cheopis. Direct pesticide control of the flea population also has been suggested to reduce flea-borne disease. However, insecticides cause a selective pressure on the flea population, leading to populations that are resistant to them. For example, the flea population in Madagascar developed resistance to DDT (dichlorodiphenyltrichloroethane), dieldrin, deltamethrin, and cyfluthrin after their widespread use.17 Further, a recent study revealed resistance of X cheopis populations to alphacypermethrin, lambda-cyhalothrin, and etofenprox, none of which were used in mass vector control, indicating that some cross-resistance mechanism between these and the extensively used insecticides may exist. With the development of widespread resistance to most pesticides, flea control in endemic areas is difficult. Insecticide targeting to fleas on rodents (eg, rodent bait containing insecticide) can allow for more targeted insecticide treatment, limiting the development of resistance.17 Recent development of a maceration protocol used to detect zoonotic pathogens in fleas in the field also will allow management with pesticides to be targeted geographically and temporally where infected vectors are located.18 Research of the interaction between vector, pathogen, and insect microbiome also should continue, as it may allow for development of biopesticides, limiting the use of chemical pesticides all together. The strategy is based on the introduction of microorganisms that can reduce vector lifespan or their ability to transmit pathogens.17
When flea-transmitted diseases do occur, treatment with antibiotics is advised. Early treatment of the plague with effective antibiotics such as streptomycin, gentamicin, tetracycline, or chloramphenicol for a minimum of 10 days is critical for survival. Additionally, patients with bubonic plague should be isolated for at least 2 days after administration of antibiotics, while patients with the pneumonic form should be isolated for 4 days into therapy to prevent the spread of disease. Prophylactic therapy for individuals who come into contact with infected individuals also is advised.4 Patients with murine typhus typically respond to doxycycline, tetracycline, or fluoroquinolones. The few cases of R felis–induced disease have responded to doxycycline. Of note, short courses of treatment of doxycycline are appropriate and safe in young children. The short (3–7 day) nature of the course limits the chances of teeth staining.14
- Bitam I, Dittmar K, Parola P, et al. Flea and flea-borne diseases. Int J Infect Dis. 2010;14:E667-E676.
- Mathison BA, Pritt BS. Laboratory identification of arthropod ectoparasites. Clin Microbiol Rev. 2014;27:48-67.
- Ligon BL. Plague: a review of its history and potential as a biological weapon. Semin Pediatr Infect Dis. 2006;17:161-170.
- Josko D. Yersinia pestis: still a plague in the 21st century. Clin Lab Sci. 2004;17:25-29.
- Plague around the world, 2010–2015. Wkly Epidemiol Rec. 2016;91:89-93.
- Sullivan K. California confirms first human case of the plague in 5 years: what to know. NBC News website. https://www.nbcnews.com/health/health-news/california-confirms-first-human-case-bubonic-plague-5-years-what-n1237306. Published August 19, 2020. Accessed August 24, 2020.
- Hinnebusch BJ, Bland DM, Bosio CF, et al. Comparative ability of Oropsylla and Xenopsylla cheopis fleas to transmit Yersinia pestis by two different mechanisms. PLOS Negl Trop Dis. 2017;11:e0005276.
- Bobrov AG, Kirillina O, Vadyvaloo V, et al. The Yersinia pestis HmsCDE regulatory system is essential for blockage of the oriental rat flea (Xenopsylla cheopis), a classic plague vector. Environ Microbiol. 2015;17:947-959.
- Darby C, Ananth SL, Tan L, et al. Identification of gmhA, a Yersina pestis gene required for flea blockage, by using a Caenorhabditis elegans biofilm system. Infect Immun. 2005;73:7236-7242.
- Eisen RJ, Dennis DT, Gage KL. The role of early-phase transmission in the spread of Yersinia pestis. J Med Entomol. 2015;52:1183-1192.
- Carniel E. Subtle genetic modifications transformed an enteropathogen into a flea-borne pathogen. Proc Natl Acad Sci U S A. 2014;111:18409-18410.
- Chouikha I, Hinnebusch BJ. Silencing urease: a key evolutionary step that facilitated the adaptation of Yersinia pestis to the flea-borne transmission route. Proc Natl Acad Sci U S A. 2014;111:18709-19714.
- Aoyagi KL, Brooks BD, Bearden SW, et al. LPS modification promotes maintenance of Yersinia pestis in fleas. Microbiology. 2015;161:628-638.
- Civen R, Ngo V. Murine typhus: an unrecognized suburban vectorborne disease. Clin Infect Dis. 2008;46:913-918.
- Eremeeva ME, Warashina WR, Sturgeon MM, et al. Rickettsia typhi and R. felis in rat fleas (Xenopsylla cheopis), Oahu, Hawaii. Emerg Infect Dis. 2018;14:1613-1615.
- Billeter SA, Gundi VAKB, Rood MP, et al. Molecular detection and identification of Bartonella species in Xenopsylla cheopis fleas (Siphonaptera: Pulicidae) collected from Rattus norvecus rats in Los Angeles, California. Appl Environ Microbiol. 2011;77:7850-7852.
- Miarinjara A, Boyer S. Current perspectives on plague vector control in Madagascar: susceptibility status of Xenopsylla cheopis to 12 insecticides. PLOS Negl Trop Dis. 2016;10:e0004414.
- Harrison GF, Scheirer JL, Melanson VR. Development and validation of an arthropod maceration protocol for zoonotic pathogen detection in mosquitoes and fleas. J Vector Ecol. 2014;40:83-89.
- Bitam I, Dittmar K, Parola P, et al. Flea and flea-borne diseases. Int J Infect Dis. 2010;14:E667-E676.
- Mathison BA, Pritt BS. Laboratory identification of arthropod ectoparasites. Clin Microbiol Rev. 2014;27:48-67.
- Ligon BL. Plague: a review of its history and potential as a biological weapon. Semin Pediatr Infect Dis. 2006;17:161-170.
- Josko D. Yersinia pestis: still a plague in the 21st century. Clin Lab Sci. 2004;17:25-29.
- Plague around the world, 2010–2015. Wkly Epidemiol Rec. 2016;91:89-93.
- Sullivan K. California confirms first human case of the plague in 5 years: what to know. NBC News website. https://www.nbcnews.com/health/health-news/california-confirms-first-human-case-bubonic-plague-5-years-what-n1237306. Published August 19, 2020. Accessed August 24, 2020.
- Hinnebusch BJ, Bland DM, Bosio CF, et al. Comparative ability of Oropsylla and Xenopsylla cheopis fleas to transmit Yersinia pestis by two different mechanisms. PLOS Negl Trop Dis. 2017;11:e0005276.
- Bobrov AG, Kirillina O, Vadyvaloo V, et al. The Yersinia pestis HmsCDE regulatory system is essential for blockage of the oriental rat flea (Xenopsylla cheopis), a classic plague vector. Environ Microbiol. 2015;17:947-959.
- Darby C, Ananth SL, Tan L, et al. Identification of gmhA, a Yersina pestis gene required for flea blockage, by using a Caenorhabditis elegans biofilm system. Infect Immun. 2005;73:7236-7242.
- Eisen RJ, Dennis DT, Gage KL. The role of early-phase transmission in the spread of Yersinia pestis. J Med Entomol. 2015;52:1183-1192.
- Carniel E. Subtle genetic modifications transformed an enteropathogen into a flea-borne pathogen. Proc Natl Acad Sci U S A. 2014;111:18409-18410.
- Chouikha I, Hinnebusch BJ. Silencing urease: a key evolutionary step that facilitated the adaptation of Yersinia pestis to the flea-borne transmission route. Proc Natl Acad Sci U S A. 2014;111:18709-19714.
- Aoyagi KL, Brooks BD, Bearden SW, et al. LPS modification promotes maintenance of Yersinia pestis in fleas. Microbiology. 2015;161:628-638.
- Civen R, Ngo V. Murine typhus: an unrecognized suburban vectorborne disease. Clin Infect Dis. 2008;46:913-918.
- Eremeeva ME, Warashina WR, Sturgeon MM, et al. Rickettsia typhi and R. felis in rat fleas (Xenopsylla cheopis), Oahu, Hawaii. Emerg Infect Dis. 2018;14:1613-1615.
- Billeter SA, Gundi VAKB, Rood MP, et al. Molecular detection and identification of Bartonella species in Xenopsylla cheopis fleas (Siphonaptera: Pulicidae) collected from Rattus norvecus rats in Los Angeles, California. Appl Environ Microbiol. 2011;77:7850-7852.
- Miarinjara A, Boyer S. Current perspectives on plague vector control in Madagascar: susceptibility status of Xenopsylla cheopis to 12 insecticides. PLOS Negl Trop Dis. 2016;10:e0004414.
- Harrison GF, Scheirer JL, Melanson VR. Development and validation of an arthropod maceration protocol for zoonotic pathogen detection in mosquitoes and fleas. J Vector Ecol. 2014;40:83-89.
Practice Points
- Xenopsylla cheopis, the oriental rat flea, is most known for carrying Yersinia pestis, the causative agent of the plague; however, it also is a vector for other bacteria, such as Rickettsia typhi, the species responsible for most cases of murine typhus.
- Despite the perception that it largely is a historical illness, modern outbreaks of plague occur in many parts of the world each year. Because fleas thrive in warm humid weather, global warming threatens the spread of the oriental rat flea and its diseases into previously unaffected parts of the world.
- There has been an effort to control oriental rat flea populations, which unfortunately has been complicated by pesticide resistance in many flea populations. It is important to continue to research the oriental rat flea and the bacterial species it carries in the hopes of finding better methods of controlling the pests and therefore decreasing illness in humans.
- Health care providers should be vigilant in identifying symptoms of flea-borne illnesses. If a patient is displaying symptoms, prompt recognition and antibiotic therapy is critical, particularly for the plague.
Which medications work best for menorrhagia?
EVIDENCE SUMMARY
A 2015 Cochrane review of the LNG-IUS for menorrhagia included 1 placebo-controlled RCT; most of the remaining 21 RCTs compared the LNG-IUS to invasive procedures such as endometrial ablation or hysterectomy.1 The placebo-controlled trial compared the LNG-IUS with placebo in 40 women on anticoagulation therapy and found a mean beneficial difference of 100 mL (95% confidence interval [CI], –116 to –83) using a subjective pictorial blood assessment chart.
Women are less likely to withdraw from LNG-IUS treatment
Four trials (379 patients) included in the Cochrane review compared LNG-IUS with combination or progesterone-only pills. All of the trials excluded women with palpable or large (> 5 cm) fibroids. In 3 trials (2 against OCPs and 1 against a 10-day course of oral progesterone), the LNG-IUS decreased MBL more than OCPs did. A fourth trial found LNG-IUS comparable to oral progesterone dosed 3 times a day from Day 5 to Day 26 of each menstrual cycle.
A recent large RCT (571 patients) that compared LNG-IUS with usual medical treatment (mefenamic acid [MFA], tranexamic acid, norethindrone, OCPs, progesterone-only pill, medroxyprogesterone acetate injection) found women significantly less likely to withdraw from LNG-IUS at 2 years (relative risk [RR] = 0.58; 95% CI, 0.49-0.70).2
Estrogen and progestin contraceptives significantly reduce bleeding
In addition to the trials in the 2015 Cochrane review comparing OCPs with LNG-IUS, a 2009 Cochrane review included a single 2-month crossover trial of 45 patients.3 This RCT compared OCPs with naproxen, MFA, and danazol to treat heavy menstrual bleeding (assessed using the alkaline haematin method).
Researchers didn’t analyze the data using intention-to-treat. No group was found to be superior. The OCP group (6 women) had a 43% reduction in MBL over baseline (no P value reported).
Tranexamic acid outperforms oral progesterone and NSAIDs but not ...
A 2018 Cochrane meta-analysis of 13 RCTs (1312 patients) of antifibrinolytics for reproductive-age women with regular heavy periods and no known underlying pathology included 4 RCTs (565 patients) that used placebo as a comparator.4 Therapy with tranexamic acid decreased blood loss by53 mL per cycle (95% CI, 44-63 mL), a 40% to 50% improvement compared with placebo. Three of the RCTs (271 patients) reported the percent of women improving on tranexamic acid as 43% to 63%, compared with 11% for placebo, resulting in an NNT of 2 to 3.
One trial (46 patients) found tranexamic acid superior to luteal phase oral progesterone, and another study (48 patients) demonstrated superiority to NSAIDs, with a mean decrease in MBL of 86 mL compared with 43 mL (P < .0027).
Continue to: On the other hand...
On the other hand, tranexamic acid compared unfavorably with LNG-IUS (1 RCT, 42 patients), showing a lower likelihood of improvement (RR = 0.43; 95% CI, 0.24-0.77). Whereas 85% of women improved with LNG-IUS, only 20% to 65% of women improved with tranexamic acid (NNT = 2 to 6).
No statistical difference was found in gastrointestinal adverse effects, headache, vaginal dryness, or dysmenorrhea.4 Only 1 thromboembolic event occurred in the 2 studies that reported this outcome, a known risk that prohibits its concomitant use with combination OCPs.
Different NSAIDs, equivalent efficacy
A 2013 Cochrane review of 18 RCTs included 8 (84 patients) that compared NSAIDs (5 MFA, 2 naproxen, 1 ibuprofen) with placebo.5 In 6 trials, NSAIDs produced a significant reduction in MBL compared with placebo, although most were crossover trials that couldn’t be compiled into the meta-analysis.
One trial (11 patients) showed a mean reduction of 124 mL (95% CI, 62-186 mL) in the MFA group. In another trial, women were less likely to report no improvement in the MFA group than in the placebo group (odds ratio [OR] = 0.08; 95% CI, 0.03-0.18). No NSAID had significantly higher efficacy than the others.
Danazol was superior to NSAIDs in a meta-analysis of 3 trials (79 patients) with a mean difference of 45 mL (95% CI, 19-71 mL), as was tranexamic acid in a single trial (48 patients) with a mean difference of 73 mL (95% CI, 22-124 mL).5 Comparisons with OCPs, oral progesterone, and an older model of LNG-IUS showed no significant differences. The most common adverse effects were gastrointestinal.
Continue to: Danazol linked to weight gain and other adverse effects
Danazol linked to weight gain and other adverse effects
A 2010 Cochrane review evaluated 9 RCTs, including 1 (66 patients) comparing danazol 200 mg with placebo that showed a significant decrease in subjectively assessed MBL in the danazol group.6 The study, which only 22 women finished, didn’t address intention-to-treat and used an unidentified scoring system. Patients also reported a significant 6.7-kg weight gain (95% CI, 1-12.4) after 3 months of treatment.
In addition to the 2013 meta-analysis showing danazol to be superior to NSAIDs, several studies6 compared danazol favorably with oral progesterone, although not all results reached significance. One study (37 patients) showed that women were more likely to rate the efficacy of danazol as moderate or high compared with progesterone (OR = 4.3; 95% CI, 1.1-17.0), but the mean difference in MBL (–36 mL; 95% CI, −102 to 31 mL) wasn’t statistically significant.
Of note, both a meta-analysis of 4 of the studies (117 patients) and another study comparing danazol with NSAIDs (20 patients) found significantly more adverse effects in the danazol group. Commonly reported adverse effects were acne, weight gain, headache, nausea, and tiredness.
RECOMMENDATIONS
A comparative effectiveness review by the Agency for Healthcare Research and Quality concluded that evidence showed efficacy for 4 primary care interventions for heavy cyclic bleeding: LNG-IUS, NSAIDs, tranexamic acid, and combination OCPs.7
The United Kingdom’s National Institute for Health Care and Excellence (NICE) recommends pharmaceutical treatment when no structural or histologic abnormality is present or when fibroids are < 3 cm in diameter.8 NICE advises considering pharmaceutical treatments in the following order: first, LNG-IUS if long-term use (at least 12 months) is anticipated; second, tranexamic acid or NSAIDs; and third, combination OCPs, norethisterone (15 mg) daily from Days 5 to 26 of the menstrual cycle, or injected long-acting progestogen.
Editor’s takeaway
I was taught to use combination OCPs as first-line treatment for menorrhagia, but better evidence supports using any of these 4: LNG-IUS, tranexamic acid, danazol, or NSAIDs. In the absence of clear evidence demonstrating differences in efficacy, I would use them in the reverse order for cost-effectiveness reasons.
1. Lethaby A, Hussain M, Rishworth JR, et al. Progesterone or progesterone-releasing intrauterine systems for heavy menstrual bleeding. Cochrane Database Syst Rev. 2015;(4):CD002126.
2. Gupta J, Kai J, Middleton L, et al. Levonorgestrel intrauterine system versus medical therapy for menorrhagia N Engl J Med. 2013;368:128-137.
3. Farquhar C, Brown J. Oral contraceptive pill for heavy menstrual bleeding. Cochrane Database Syst Rev. 2009;(4):CD000154.
4. Bryant-Smith AC, Lethaby A, Farquhar C, et al. Antifibrinolytics for heavy menstrual bleeding. Cochrane Database Syst Rev. 2018;(4):CD000249.
5. Lethaby A, Duckitt K, Farquhar C. Non-steroidal anti-inflammatory drugs for heavy menstrual bleeding. Cochrane Database Syst Rev. 2013;(1):CD000400.
6. Beaumont HH, Augood C, Duckitt K, et al. Danazol for heavy menstrual bleeding. Cochrane Database Syst Rev. 2010;(1):CD00107.
7. Hartmann KE, Jerome RN, Lindegren ML, et al. Primary Care Management of Abnormal Uterine Bleeding. Comparative Effectiveness Review No. 96 (AHRQ Publication No. 13-EHC025-EF). Rockville, MD: Agency for Healthcare Research and Quality; 2013. https://effectivehealthcare.ahrq.gov/topics/abnormal-uterine-bleeding. Accessed August 25, 2020.
8. National Institute for Health Care and Excellence (NICE). Heavy menstrual bleeding: assessment and management. NICE Guideline NG88; 2018. www.nice.org.uk/guidance/ng88. Accessed August 25, 2020.
EVIDENCE SUMMARY
A 2015 Cochrane review of the LNG-IUS for menorrhagia included 1 placebo-controlled RCT; most of the remaining 21 RCTs compared the LNG-IUS to invasive procedures such as endometrial ablation or hysterectomy.1 The placebo-controlled trial compared the LNG-IUS with placebo in 40 women on anticoagulation therapy and found a mean beneficial difference of 100 mL (95% confidence interval [CI], –116 to –83) using a subjective pictorial blood assessment chart.
Women are less likely to withdraw from LNG-IUS treatment
Four trials (379 patients) included in the Cochrane review compared LNG-IUS with combination or progesterone-only pills. All of the trials excluded women with palpable or large (> 5 cm) fibroids. In 3 trials (2 against OCPs and 1 against a 10-day course of oral progesterone), the LNG-IUS decreased MBL more than OCPs did. A fourth trial found LNG-IUS comparable to oral progesterone dosed 3 times a day from Day 5 to Day 26 of each menstrual cycle.
A recent large RCT (571 patients) that compared LNG-IUS with usual medical treatment (mefenamic acid [MFA], tranexamic acid, norethindrone, OCPs, progesterone-only pill, medroxyprogesterone acetate injection) found women significantly less likely to withdraw from LNG-IUS at 2 years (relative risk [RR] = 0.58; 95% CI, 0.49-0.70).2
Estrogen and progestin contraceptives significantly reduce bleeding
In addition to the trials in the 2015 Cochrane review comparing OCPs with LNG-IUS, a 2009 Cochrane review included a single 2-month crossover trial of 45 patients.3 This RCT compared OCPs with naproxen, MFA, and danazol to treat heavy menstrual bleeding (assessed using the alkaline haematin method).
Researchers didn’t analyze the data using intention-to-treat. No group was found to be superior. The OCP group (6 women) had a 43% reduction in MBL over baseline (no P value reported).
Tranexamic acid outperforms oral progesterone and NSAIDs but not ...
A 2018 Cochrane meta-analysis of 13 RCTs (1312 patients) of antifibrinolytics for reproductive-age women with regular heavy periods and no known underlying pathology included 4 RCTs (565 patients) that used placebo as a comparator.4 Therapy with tranexamic acid decreased blood loss by53 mL per cycle (95% CI, 44-63 mL), a 40% to 50% improvement compared with placebo. Three of the RCTs (271 patients) reported the percent of women improving on tranexamic acid as 43% to 63%, compared with 11% for placebo, resulting in an NNT of 2 to 3.
One trial (46 patients) found tranexamic acid superior to luteal phase oral progesterone, and another study (48 patients) demonstrated superiority to NSAIDs, with a mean decrease in MBL of 86 mL compared with 43 mL (P < .0027).
Continue to: On the other hand...
On the other hand, tranexamic acid compared unfavorably with LNG-IUS (1 RCT, 42 patients), showing a lower likelihood of improvement (RR = 0.43; 95% CI, 0.24-0.77). Whereas 85% of women improved with LNG-IUS, only 20% to 65% of women improved with tranexamic acid (NNT = 2 to 6).
No statistical difference was found in gastrointestinal adverse effects, headache, vaginal dryness, or dysmenorrhea.4 Only 1 thromboembolic event occurred in the 2 studies that reported this outcome, a known risk that prohibits its concomitant use with combination OCPs.
Different NSAIDs, equivalent efficacy
A 2013 Cochrane review of 18 RCTs included 8 (84 patients) that compared NSAIDs (5 MFA, 2 naproxen, 1 ibuprofen) with placebo.5 In 6 trials, NSAIDs produced a significant reduction in MBL compared with placebo, although most were crossover trials that couldn’t be compiled into the meta-analysis.
One trial (11 patients) showed a mean reduction of 124 mL (95% CI, 62-186 mL) in the MFA group. In another trial, women were less likely to report no improvement in the MFA group than in the placebo group (odds ratio [OR] = 0.08; 95% CI, 0.03-0.18). No NSAID had significantly higher efficacy than the others.
Danazol was superior to NSAIDs in a meta-analysis of 3 trials (79 patients) with a mean difference of 45 mL (95% CI, 19-71 mL), as was tranexamic acid in a single trial (48 patients) with a mean difference of 73 mL (95% CI, 22-124 mL).5 Comparisons with OCPs, oral progesterone, and an older model of LNG-IUS showed no significant differences. The most common adverse effects were gastrointestinal.
Continue to: Danazol linked to weight gain and other adverse effects
Danazol linked to weight gain and other adverse effects
A 2010 Cochrane review evaluated 9 RCTs, including 1 (66 patients) comparing danazol 200 mg with placebo that showed a significant decrease in subjectively assessed MBL in the danazol group.6 The study, which only 22 women finished, didn’t address intention-to-treat and used an unidentified scoring system. Patients also reported a significant 6.7-kg weight gain (95% CI, 1-12.4) after 3 months of treatment.
In addition to the 2013 meta-analysis showing danazol to be superior to NSAIDs, several studies6 compared danazol favorably with oral progesterone, although not all results reached significance. One study (37 patients) showed that women were more likely to rate the efficacy of danazol as moderate or high compared with progesterone (OR = 4.3; 95% CI, 1.1-17.0), but the mean difference in MBL (–36 mL; 95% CI, −102 to 31 mL) wasn’t statistically significant.
Of note, both a meta-analysis of 4 of the studies (117 patients) and another study comparing danazol with NSAIDs (20 patients) found significantly more adverse effects in the danazol group. Commonly reported adverse effects were acne, weight gain, headache, nausea, and tiredness.
RECOMMENDATIONS
A comparative effectiveness review by the Agency for Healthcare Research and Quality concluded that evidence showed efficacy for 4 primary care interventions for heavy cyclic bleeding: LNG-IUS, NSAIDs, tranexamic acid, and combination OCPs.7
The United Kingdom’s National Institute for Health Care and Excellence (NICE) recommends pharmaceutical treatment when no structural or histologic abnormality is present or when fibroids are < 3 cm in diameter.8 NICE advises considering pharmaceutical treatments in the following order: first, LNG-IUS if long-term use (at least 12 months) is anticipated; second, tranexamic acid or NSAIDs; and third, combination OCPs, norethisterone (15 mg) daily from Days 5 to 26 of the menstrual cycle, or injected long-acting progestogen.
Editor’s takeaway
I was taught to use combination OCPs as first-line treatment for menorrhagia, but better evidence supports using any of these 4: LNG-IUS, tranexamic acid, danazol, or NSAIDs. In the absence of clear evidence demonstrating differences in efficacy, I would use them in the reverse order for cost-effectiveness reasons.
EVIDENCE SUMMARY
A 2015 Cochrane review of the LNG-IUS for menorrhagia included 1 placebo-controlled RCT; most of the remaining 21 RCTs compared the LNG-IUS to invasive procedures such as endometrial ablation or hysterectomy.1 The placebo-controlled trial compared the LNG-IUS with placebo in 40 women on anticoagulation therapy and found a mean beneficial difference of 100 mL (95% confidence interval [CI], –116 to –83) using a subjective pictorial blood assessment chart.
Women are less likely to withdraw from LNG-IUS treatment
Four trials (379 patients) included in the Cochrane review compared LNG-IUS with combination or progesterone-only pills. All of the trials excluded women with palpable or large (> 5 cm) fibroids. In 3 trials (2 against OCPs and 1 against a 10-day course of oral progesterone), the LNG-IUS decreased MBL more than OCPs did. A fourth trial found LNG-IUS comparable to oral progesterone dosed 3 times a day from Day 5 to Day 26 of each menstrual cycle.
A recent large RCT (571 patients) that compared LNG-IUS with usual medical treatment (mefenamic acid [MFA], tranexamic acid, norethindrone, OCPs, progesterone-only pill, medroxyprogesterone acetate injection) found women significantly less likely to withdraw from LNG-IUS at 2 years (relative risk [RR] = 0.58; 95% CI, 0.49-0.70).2
Estrogen and progestin contraceptives significantly reduce bleeding
In addition to the trials in the 2015 Cochrane review comparing OCPs with LNG-IUS, a 2009 Cochrane review included a single 2-month crossover trial of 45 patients.3 This RCT compared OCPs with naproxen, MFA, and danazol to treat heavy menstrual bleeding (assessed using the alkaline haematin method).
Researchers didn’t analyze the data using intention-to-treat. No group was found to be superior. The OCP group (6 women) had a 43% reduction in MBL over baseline (no P value reported).
Tranexamic acid outperforms oral progesterone and NSAIDs but not ...
A 2018 Cochrane meta-analysis of 13 RCTs (1312 patients) of antifibrinolytics for reproductive-age women with regular heavy periods and no known underlying pathology included 4 RCTs (565 patients) that used placebo as a comparator.4 Therapy with tranexamic acid decreased blood loss by53 mL per cycle (95% CI, 44-63 mL), a 40% to 50% improvement compared with placebo. Three of the RCTs (271 patients) reported the percent of women improving on tranexamic acid as 43% to 63%, compared with 11% for placebo, resulting in an NNT of 2 to 3.
One trial (46 patients) found tranexamic acid superior to luteal phase oral progesterone, and another study (48 patients) demonstrated superiority to NSAIDs, with a mean decrease in MBL of 86 mL compared with 43 mL (P < .0027).
Continue to: On the other hand...
On the other hand, tranexamic acid compared unfavorably with LNG-IUS (1 RCT, 42 patients), showing a lower likelihood of improvement (RR = 0.43; 95% CI, 0.24-0.77). Whereas 85% of women improved with LNG-IUS, only 20% to 65% of women improved with tranexamic acid (NNT = 2 to 6).
No statistical difference was found in gastrointestinal adverse effects, headache, vaginal dryness, or dysmenorrhea.4 Only 1 thromboembolic event occurred in the 2 studies that reported this outcome, a known risk that prohibits its concomitant use with combination OCPs.
Different NSAIDs, equivalent efficacy
A 2013 Cochrane review of 18 RCTs included 8 (84 patients) that compared NSAIDs (5 MFA, 2 naproxen, 1 ibuprofen) with placebo.5 In 6 trials, NSAIDs produced a significant reduction in MBL compared with placebo, although most were crossover trials that couldn’t be compiled into the meta-analysis.
One trial (11 patients) showed a mean reduction of 124 mL (95% CI, 62-186 mL) in the MFA group. In another trial, women were less likely to report no improvement in the MFA group than in the placebo group (odds ratio [OR] = 0.08; 95% CI, 0.03-0.18). No NSAID had significantly higher efficacy than the others.
Danazol was superior to NSAIDs in a meta-analysis of 3 trials (79 patients) with a mean difference of 45 mL (95% CI, 19-71 mL), as was tranexamic acid in a single trial (48 patients) with a mean difference of 73 mL (95% CI, 22-124 mL).5 Comparisons with OCPs, oral progesterone, and an older model of LNG-IUS showed no significant differences. The most common adverse effects were gastrointestinal.
Continue to: Danazol linked to weight gain and other adverse effects
Danazol linked to weight gain and other adverse effects
A 2010 Cochrane review evaluated 9 RCTs, including 1 (66 patients) comparing danazol 200 mg with placebo that showed a significant decrease in subjectively assessed MBL in the danazol group.6 The study, which only 22 women finished, didn’t address intention-to-treat and used an unidentified scoring system. Patients also reported a significant 6.7-kg weight gain (95% CI, 1-12.4) after 3 months of treatment.
In addition to the 2013 meta-analysis showing danazol to be superior to NSAIDs, several studies6 compared danazol favorably with oral progesterone, although not all results reached significance. One study (37 patients) showed that women were more likely to rate the efficacy of danazol as moderate or high compared with progesterone (OR = 4.3; 95% CI, 1.1-17.0), but the mean difference in MBL (–36 mL; 95% CI, −102 to 31 mL) wasn’t statistically significant.
Of note, both a meta-analysis of 4 of the studies (117 patients) and another study comparing danazol with NSAIDs (20 patients) found significantly more adverse effects in the danazol group. Commonly reported adverse effects were acne, weight gain, headache, nausea, and tiredness.
RECOMMENDATIONS
A comparative effectiveness review by the Agency for Healthcare Research and Quality concluded that evidence showed efficacy for 4 primary care interventions for heavy cyclic bleeding: LNG-IUS, NSAIDs, tranexamic acid, and combination OCPs.7
The United Kingdom’s National Institute for Health Care and Excellence (NICE) recommends pharmaceutical treatment when no structural or histologic abnormality is present or when fibroids are < 3 cm in diameter.8 NICE advises considering pharmaceutical treatments in the following order: first, LNG-IUS if long-term use (at least 12 months) is anticipated; second, tranexamic acid or NSAIDs; and third, combination OCPs, norethisterone (15 mg) daily from Days 5 to 26 of the menstrual cycle, or injected long-acting progestogen.
Editor’s takeaway
I was taught to use combination OCPs as first-line treatment for menorrhagia, but better evidence supports using any of these 4: LNG-IUS, tranexamic acid, danazol, or NSAIDs. In the absence of clear evidence demonstrating differences in efficacy, I would use them in the reverse order for cost-effectiveness reasons.
1. Lethaby A, Hussain M, Rishworth JR, et al. Progesterone or progesterone-releasing intrauterine systems for heavy menstrual bleeding. Cochrane Database Syst Rev. 2015;(4):CD002126.
2. Gupta J, Kai J, Middleton L, et al. Levonorgestrel intrauterine system versus medical therapy for menorrhagia N Engl J Med. 2013;368:128-137.
3. Farquhar C, Brown J. Oral contraceptive pill for heavy menstrual bleeding. Cochrane Database Syst Rev. 2009;(4):CD000154.
4. Bryant-Smith AC, Lethaby A, Farquhar C, et al. Antifibrinolytics for heavy menstrual bleeding. Cochrane Database Syst Rev. 2018;(4):CD000249.
5. Lethaby A, Duckitt K, Farquhar C. Non-steroidal anti-inflammatory drugs for heavy menstrual bleeding. Cochrane Database Syst Rev. 2013;(1):CD000400.
6. Beaumont HH, Augood C, Duckitt K, et al. Danazol for heavy menstrual bleeding. Cochrane Database Syst Rev. 2010;(1):CD00107.
7. Hartmann KE, Jerome RN, Lindegren ML, et al. Primary Care Management of Abnormal Uterine Bleeding. Comparative Effectiveness Review No. 96 (AHRQ Publication No. 13-EHC025-EF). Rockville, MD: Agency for Healthcare Research and Quality; 2013. https://effectivehealthcare.ahrq.gov/topics/abnormal-uterine-bleeding. Accessed August 25, 2020.
8. National Institute for Health Care and Excellence (NICE). Heavy menstrual bleeding: assessment and management. NICE Guideline NG88; 2018. www.nice.org.uk/guidance/ng88. Accessed August 25, 2020.
1. Lethaby A, Hussain M, Rishworth JR, et al. Progesterone or progesterone-releasing intrauterine systems for heavy menstrual bleeding. Cochrane Database Syst Rev. 2015;(4):CD002126.
2. Gupta J, Kai J, Middleton L, et al. Levonorgestrel intrauterine system versus medical therapy for menorrhagia N Engl J Med. 2013;368:128-137.
3. Farquhar C, Brown J. Oral contraceptive pill for heavy menstrual bleeding. Cochrane Database Syst Rev. 2009;(4):CD000154.
4. Bryant-Smith AC, Lethaby A, Farquhar C, et al. Antifibrinolytics for heavy menstrual bleeding. Cochrane Database Syst Rev. 2018;(4):CD000249.
5. Lethaby A, Duckitt K, Farquhar C. Non-steroidal anti-inflammatory drugs for heavy menstrual bleeding. Cochrane Database Syst Rev. 2013;(1):CD000400.
6. Beaumont HH, Augood C, Duckitt K, et al. Danazol for heavy menstrual bleeding. Cochrane Database Syst Rev. 2010;(1):CD00107.
7. Hartmann KE, Jerome RN, Lindegren ML, et al. Primary Care Management of Abnormal Uterine Bleeding. Comparative Effectiveness Review No. 96 (AHRQ Publication No. 13-EHC025-EF). Rockville, MD: Agency for Healthcare Research and Quality; 2013. https://effectivehealthcare.ahrq.gov/topics/abnormal-uterine-bleeding. Accessed August 25, 2020.
8. National Institute for Health Care and Excellence (NICE). Heavy menstrual bleeding: assessment and management. NICE Guideline NG88; 2018. www.nice.org.uk/guidance/ng88. Accessed August 25, 2020.
EVIDENCE-BASED ANSWER:
Four medications have been shown to reduce menstrual blood loss (MBL) significantly in placebo-controlled randomized controlled trials (RCTs): the levonorgestrel-releasing intrauterine system (LNG-IUS), tranexamic acid, nonsteroidal anti-inflammatory drugs (NSAIDs), and danazol, a synthetic steroid (strength of recommendation: A, meta-analyses of RCTs).
A single trial showed that the LNG-IUS reduced MBL by about 100 mL, compared with placebo. In a meta-analysis of 4 placebo-controlled RCTs, tranexamic acid reduced MBL by about 53 mL, roughly a 40% to 50% decrease. The 8 NSAID trials (5 mefenamic acid, 2 naproxen, 1 ibuprofen) demonstrated effectiveness, but the effect size is difficult to quantify. The single danazol RCT used a subjective scoring system without reporting MBL.
No studies compared all effective medical therapies against one another. In head-to-head comparisons, women were more likely to experience improvement with the LNG-IUS than with tranexamic acid (number needed to treat [NNT] = 2 to 6). Both treatments are superior to NSAIDs. Danazol is also more efficacious than NSAIDs, but its use is limited by its adverse effects, including teratogenicity.
No placebo-controlled trials have studied oral contraceptive pills (OCPs) or oral progesterone to treat menorrhagia. However, multiple comparative RCTs have demonstrated that these commonly prescribed medications significantly decrease MBL. Trials have shown the reduction to be inferior to LNG-IUS and danazol and equivalent to NSAIDs.
Management of Classic Ulcerative Pyoderma Gangrenosum
Pyoderma gangrenosum (PG) is a rare, chronic, ulcerative, neutrophilic dermatosis of unclear etiology. Large, multicentered, randomized controlled trials (RCTs) are challenging due to the rarity of PG and the lack of a diagnostic confirmatory test; therefore, evidence-based guidelines for diagnosis and treatment are not well established. Current management of PG primarily is guided by case series, small clinical trials, and expert opinion.1-4 We conducted a survey of expert medical dermatologists to highlight best practices in diagnostic and therapeutic approaches to PG.
Methods
The Society of Dermatology Hospitalists (SDH) Scientific Task Force gathered expert opinions from members of the SDH and Rheumatologic Dermatology Society (RDS) regarding PG workup and treatment through an online survey of 15 items (eTable 1). Subscribers of the SDH and RDS LISTSERVs were invited via email to participate in the survey from January 2016 to February 2016. Anonymous survey responses were collected and collated using SurveyMonkey. The survey results identified expert recommendations for evaluation, diagnosis, and treatment of PG and are reported as the sum of the percentage of respondents who answered always (almost 100% of the time) or often (more than half the time) following a particular course of action. A subanalysis was performed defining 2 groups of respondents based on the number of cases of PG treated per year (≥10 vs <10). Survey responses between each group were compared using χ2 analysis with statistical significance set at P=.05.
Results
Fifty-one respondents completed the survey out of 140 surveyed (36% response rate). All respondents were dermatologists, and 96% (49/51) were affiliated with an academic institution. Among the respondents, the number of PG cases managed per year ranged from 2 to 35.
Respondents consistently ordered skin biopsies (92% [47/51]) and tissue cultures (90% [46/51]), as well as certain ancillary tests, including complete blood cell count (96% [49/51]), complete metabolic panel (86% [44/51]), serum protein electrophoresis (76% [39/51]), and hepatitis panel (71% [36/51]). Other frequently ordered studies were rheumatoid factor (69% [35/51]), antinuclear antibodies (67% [34/51]), and antineutrophilic antibodies (65% [33/51]). Respondents frequently ordered erythrocyte sedimentation rate (59% [30/51]), C-reactive protein (55% [28/51]), cryoglobulins (53% [27/51]), urine protein electrophoresis (53% [27/51]), hypercoagulability workup (49% [25/51]), and serum immunofixation test (49% [25/51]). Human immunodeficiency virus testing (43% [22/51]), chest radiograph (41% [21/51]), colonoscopy (41% [21/51]) and referral to other specialties for workup—gastroenterology (38% [19/51]), hematology/oncology (14% [7/51]), and rheumatology (10% [5/51])—were less frequently ordered (eTable 2).
Systemic corticosteroids were reported as first-line therapy by most respondents (94% [48/51]), followed by topical immunomodulatory therapies (63% [32/51]). Topical corticosteroids (75% [38/51]) were the most common first-line topical agents. Thirty-nine percent of respondents (20/51) prescribed topical calcineurin inhibitors as first-line topical therapy. Additional therapies frequently used included systemic cyclosporine (47% [24/51]), antineutrophilic agents (41% [21/51]), and biologic agents (37% [19/51]). Fifty-seven percent of respondents (29/51) supported using combination topical and systemic therapy (Table).
A wide variety of wound care practices were reported in the management of PG. Seventy-six percent of respondents (39/51) favored petroleum-impregnated gauze, 69% (35/51) used nonadhesive dressings, and 43% (22/51) added antimicrobial therapy for PG wound care (eTable 3). In the subanalysis, there were no significant differences in the majority of answer responses in patients treating 10 or more PG cases per year vs fewer than 10 PG cases, except with regard to the practice of combination therapy. Those treating more than 10 cases of PG per year more frequently reported use of combination therapies compared to respondents treating fewer than 10 cases (P=.04).
Comment
Skin biopsies and tissue cultures were strongly recommended (>90% survey respondents) for the initial evaluation of lesions suspected to be PG to evaluate for typical histopathologic changes that appear early in the disease, to rule out PG mimickers such as infectious or vascular causes, and to prevent the detrimental effects of inappropriate treatment and delayed diagnosis.5
Suspected PG warrants a reasonable search for related conditions because more than 50% of PG cases are associated with comorbidities such as rheumatoid arthritis, inflammatory bowel disease, and hematologic disease/malignancy.6,7 A complete blood cell count and comprehensive metabolic panel were recommended by most respondents, aiding in the preliminary screening for hematologic and infectious causes as well as detecting liver and kidney dysfunction associated with systemic conditions. Additionally, exclusion of infection or malignancy may be particularly important if the patient will undergo systemic immunosuppression. In challenging PG cases when initial findings are inconclusive and the clinical presentation does not direct workup (eg, colonoscopy to evaluate gastrointestinal tract symptoms), serum protein electrophoresis, hepatitis panel, rheumatoid factor, antinuclear antibodies, and antineutrophilic antibody tests also were frequently ordered by respondents to further evaluate for underlying or associated conditions.
This consensus regarding skin biopsies and certain ancillary tests is consistent with the proposed diagnostic criteria for classic ulcerative PG in which the absence or exclusion of other relevant causes of cutaneous ulcers is required based on the criteria.8 The importance of ensuring an accurate diagnosis is paramount, as a 10% misdiagnosis rate has been documented in the literature.5
Importantly, a stepwise diagnostic workup for PG is proposed based on survey results, which may limit unnecessary testing and the associated costs to the health care system (Figure 1). Selection of additional testing is guided by initial test results and features of the patient’s clinical presentation, including age, review of systems, and associated comorbidities. Available data suggest that underlying inflammatory bowel disease is more frequent in PG patients who are younger than 65 years, whereas those who are 65 years and older are more likely to have inflammatory arthritis, cancer, or an underlying hematologic disorder.9
Treatment of PG should address both the inflammatory and wound components of the disease (Figure 2).7 In our survey results, systemic corticosteroids were identified as an important first-line therapy supported by reasonable evidence and were favored for their rapid response and minimal cost.1,10,11 Many respondents endorsed the use of systemic therapy in combination with topical steroids or calcineurin inhibitors. Combination therapy may provide more immediate control of rapidly progressing disease while minimizing adverse effects of long-term systemic corticosteroid use. A survey of German wound experts similarly endorsed frequent use of topical calcineurin inhibitors and combination systemic and topical glucocorticoid therapy as common therapeutic approaches.1
Importantly, treatments may vary depending on patient characteristics, comorbidities, and underlying disease, which underscores the need for individualized treatment approaches. Alternative first-line systemic treatments favored by respondents were cyclosporine, biologic medications, and antineutrophilic agents such as dapsone. Cyclosporine has demonstrated comparable efficacy to systemic glucocorticoids in one RCT and is considered an important steroid-sparing alternative for PG treatment.2 Biologic agents, especially tumor necrosis factor inhibitors, may be effective in treating cases of refractory PG or for concomitant inflammatory bowel disease management, as demonstrated by a small RCT documenting improvement of PG following infliximab infusion.3
Respondents strongly recommended petrolatum-impregnated gauze and other nonadhesive dressings, including alginate and hydrocolloid dressings, as part of PG wound care. Topical antimicrobials and compression stockings also were recommended by respondents. These practices aim to promote moist environments for healing, avoid maceration, prevent superinfection, optimize wound healing, and minimize damage from adhesive injury.12 Wound debridement and grafting generally were not recommended. However, pathergy is not a universal phenomenon in PG, and wounds that are no longer in the inflammatory phase may benefit from gentle debridement of necrotic tissue and/or grafting in select cases.10
Conclusion
An approach to modifying PG management based on clinical presentation and the practice of combination therapy with multiple systemic agents in refractory PG cases was not addressed in our survey. The low response rate is a limitation; however, the opinions of 51 medical dermatologist experts who regularly manage PG (in contrast to papers based on individualized clinical experience) can provide important clinical guidance until more scientific evidence is established.
Acknowledgments
We would like to thank the SDH and RDS membership for their participation in this survey. We especially acknowledge the other members of the SDH Scientific Task Force for their feedback: Misha Rosenbach, MD (Philadelphia, Pennsylvania); Robert G. Micheletti, MD (Philadelphia, Pennsylvania); Karolyn Wanat, MD (Milwaukee, Wisconsin); Amy Chen, MD (Cromwell, Connecticut); and A. Rambi Cardones, MD (Durham, North Carolina).
- Al Ghazal P, Dissemond J. Therapy of pyoderma gangrenosum in Germany: results of a survey among wound experts. J Dtsch Dermatol Ges . 2015;13:317-324.
- Ormerod AD, Thomas KS, Craig FE, et al. Comparison of the two most commonly used treatments for pyoderma gangrenosum: results of the STOP GAP randomised controlled trial. BMJ. 2015;350:h2958.
- Brooklyn TN, Dunnill MG, Shetty A, et al. Infliximab for the treatment of pyoderma gangrenosum: a randomised, double blind, placebo controlled trial. Gut. 2006;55:505-509.
- Al Ghazal P, Klode J, Dissemond J. Diagnostic criteria for pyoderma gangrenosum: results of a survey among dermatologic wound experts in Germany. J Dtsch Dermatol Ges. 2014;12:1129-1131.
- Weenig RH, Davis MD, Dahl PR, et al. Skin ulcers misdiagnosed as pyoderma gangrenosum. N Engl J Med. 2002;347:1412-1418.
- Powell FC, Su WP, Perry HO. Pyoderma gangrenosum: classification and management. J Am Acad Dermatol. 1996;34:395-409.
- Bennett ML, Jackson JM, Jorizzo JL, et al. Pyoderma gangrenosum: a comparison of typical and atypical forms with an emphasis on time to remission. case review of 86 patients from 2 institutions. Medicine. 2000;79:37-46.
- Su WP, Davis MD, Weening RH, et al. Pyoderma gangrenosum: clinicopathologic correlation and proposed diagnostic criteria. Int J Dermatol. 2004;43:790-800.
- Aschyan H, Butler DC, Nelson CA, et al. The association of age with clinical presentation and comorbidities of pyoderma gangrenosum. JAMA Dermatol. 2018;154:409-413.
- Binus AM, Qureshi AA, Li VW, et al. Pyoderma gangrenosum: a retrospective review of patient characteristics, comorbidities and therapy in 103 patients. Br J Dermatol. 2011;165:1244-1250.
- Reichrath J, Bens G, Bonowitz A, et al. Treatment recommendations for pyoderma gangrenosum: an evidence-based review of the literature based on more than 350 patients. J Am Acad Dermatol. 2005;53:273-283.
- Miller J, Yentzer BA, Clark A, et al. Pyoderma gangrenosum: a review and update on new therapies. J Am Acad Dermatol. 2010;62:646-654.
Pyoderma gangrenosum (PG) is a rare, chronic, ulcerative, neutrophilic dermatosis of unclear etiology. Large, multicentered, randomized controlled trials (RCTs) are challenging due to the rarity of PG and the lack of a diagnostic confirmatory test; therefore, evidence-based guidelines for diagnosis and treatment are not well established. Current management of PG primarily is guided by case series, small clinical trials, and expert opinion.1-4 We conducted a survey of expert medical dermatologists to highlight best practices in diagnostic and therapeutic approaches to PG.
Methods
The Society of Dermatology Hospitalists (SDH) Scientific Task Force gathered expert opinions from members of the SDH and Rheumatologic Dermatology Society (RDS) regarding PG workup and treatment through an online survey of 15 items (eTable 1). Subscribers of the SDH and RDS LISTSERVs were invited via email to participate in the survey from January 2016 to February 2016. Anonymous survey responses were collected and collated using SurveyMonkey. The survey results identified expert recommendations for evaluation, diagnosis, and treatment of PG and are reported as the sum of the percentage of respondents who answered always (almost 100% of the time) or often (more than half the time) following a particular course of action. A subanalysis was performed defining 2 groups of respondents based on the number of cases of PG treated per year (≥10 vs <10). Survey responses between each group were compared using χ2 analysis with statistical significance set at P=.05.
Results
Fifty-one respondents completed the survey out of 140 surveyed (36% response rate). All respondents were dermatologists, and 96% (49/51) were affiliated with an academic institution. Among the respondents, the number of PG cases managed per year ranged from 2 to 35.
Respondents consistently ordered skin biopsies (92% [47/51]) and tissue cultures (90% [46/51]), as well as certain ancillary tests, including complete blood cell count (96% [49/51]), complete metabolic panel (86% [44/51]), serum protein electrophoresis (76% [39/51]), and hepatitis panel (71% [36/51]). Other frequently ordered studies were rheumatoid factor (69% [35/51]), antinuclear antibodies (67% [34/51]), and antineutrophilic antibodies (65% [33/51]). Respondents frequently ordered erythrocyte sedimentation rate (59% [30/51]), C-reactive protein (55% [28/51]), cryoglobulins (53% [27/51]), urine protein electrophoresis (53% [27/51]), hypercoagulability workup (49% [25/51]), and serum immunofixation test (49% [25/51]). Human immunodeficiency virus testing (43% [22/51]), chest radiograph (41% [21/51]), colonoscopy (41% [21/51]) and referral to other specialties for workup—gastroenterology (38% [19/51]), hematology/oncology (14% [7/51]), and rheumatology (10% [5/51])—were less frequently ordered (eTable 2).
Systemic corticosteroids were reported as first-line therapy by most respondents (94% [48/51]), followed by topical immunomodulatory therapies (63% [32/51]). Topical corticosteroids (75% [38/51]) were the most common first-line topical agents. Thirty-nine percent of respondents (20/51) prescribed topical calcineurin inhibitors as first-line topical therapy. Additional therapies frequently used included systemic cyclosporine (47% [24/51]), antineutrophilic agents (41% [21/51]), and biologic agents (37% [19/51]). Fifty-seven percent of respondents (29/51) supported using combination topical and systemic therapy (Table).
A wide variety of wound care practices were reported in the management of PG. Seventy-six percent of respondents (39/51) favored petroleum-impregnated gauze, 69% (35/51) used nonadhesive dressings, and 43% (22/51) added antimicrobial therapy for PG wound care (eTable 3). In the subanalysis, there were no significant differences in the majority of answer responses in patients treating 10 or more PG cases per year vs fewer than 10 PG cases, except with regard to the practice of combination therapy. Those treating more than 10 cases of PG per year more frequently reported use of combination therapies compared to respondents treating fewer than 10 cases (P=.04).
Comment
Skin biopsies and tissue cultures were strongly recommended (>90% survey respondents) for the initial evaluation of lesions suspected to be PG to evaluate for typical histopathologic changes that appear early in the disease, to rule out PG mimickers such as infectious or vascular causes, and to prevent the detrimental effects of inappropriate treatment and delayed diagnosis.5
Suspected PG warrants a reasonable search for related conditions because more than 50% of PG cases are associated with comorbidities such as rheumatoid arthritis, inflammatory bowel disease, and hematologic disease/malignancy.6,7 A complete blood cell count and comprehensive metabolic panel were recommended by most respondents, aiding in the preliminary screening for hematologic and infectious causes as well as detecting liver and kidney dysfunction associated with systemic conditions. Additionally, exclusion of infection or malignancy may be particularly important if the patient will undergo systemic immunosuppression. In challenging PG cases when initial findings are inconclusive and the clinical presentation does not direct workup (eg, colonoscopy to evaluate gastrointestinal tract symptoms), serum protein electrophoresis, hepatitis panel, rheumatoid factor, antinuclear antibodies, and antineutrophilic antibody tests also were frequently ordered by respondents to further evaluate for underlying or associated conditions.
This consensus regarding skin biopsies and certain ancillary tests is consistent with the proposed diagnostic criteria for classic ulcerative PG in which the absence or exclusion of other relevant causes of cutaneous ulcers is required based on the criteria.8 The importance of ensuring an accurate diagnosis is paramount, as a 10% misdiagnosis rate has been documented in the literature.5
Importantly, a stepwise diagnostic workup for PG is proposed based on survey results, which may limit unnecessary testing and the associated costs to the health care system (Figure 1). Selection of additional testing is guided by initial test results and features of the patient’s clinical presentation, including age, review of systems, and associated comorbidities. Available data suggest that underlying inflammatory bowel disease is more frequent in PG patients who are younger than 65 years, whereas those who are 65 years and older are more likely to have inflammatory arthritis, cancer, or an underlying hematologic disorder.9
Treatment of PG should address both the inflammatory and wound components of the disease (Figure 2).7 In our survey results, systemic corticosteroids were identified as an important first-line therapy supported by reasonable evidence and were favored for their rapid response and minimal cost.1,10,11 Many respondents endorsed the use of systemic therapy in combination with topical steroids or calcineurin inhibitors. Combination therapy may provide more immediate control of rapidly progressing disease while minimizing adverse effects of long-term systemic corticosteroid use. A survey of German wound experts similarly endorsed frequent use of topical calcineurin inhibitors and combination systemic and topical glucocorticoid therapy as common therapeutic approaches.1
Importantly, treatments may vary depending on patient characteristics, comorbidities, and underlying disease, which underscores the need for individualized treatment approaches. Alternative first-line systemic treatments favored by respondents were cyclosporine, biologic medications, and antineutrophilic agents such as dapsone. Cyclosporine has demonstrated comparable efficacy to systemic glucocorticoids in one RCT and is considered an important steroid-sparing alternative for PG treatment.2 Biologic agents, especially tumor necrosis factor inhibitors, may be effective in treating cases of refractory PG or for concomitant inflammatory bowel disease management, as demonstrated by a small RCT documenting improvement of PG following infliximab infusion.3
Respondents strongly recommended petrolatum-impregnated gauze and other nonadhesive dressings, including alginate and hydrocolloid dressings, as part of PG wound care. Topical antimicrobials and compression stockings also were recommended by respondents. These practices aim to promote moist environments for healing, avoid maceration, prevent superinfection, optimize wound healing, and minimize damage from adhesive injury.12 Wound debridement and grafting generally were not recommended. However, pathergy is not a universal phenomenon in PG, and wounds that are no longer in the inflammatory phase may benefit from gentle debridement of necrotic tissue and/or grafting in select cases.10
Conclusion
An approach to modifying PG management based on clinical presentation and the practice of combination therapy with multiple systemic agents in refractory PG cases was not addressed in our survey. The low response rate is a limitation; however, the opinions of 51 medical dermatologist experts who regularly manage PG (in contrast to papers based on individualized clinical experience) can provide important clinical guidance until more scientific evidence is established.
Acknowledgments
We would like to thank the SDH and RDS membership for their participation in this survey. We especially acknowledge the other members of the SDH Scientific Task Force for their feedback: Misha Rosenbach, MD (Philadelphia, Pennsylvania); Robert G. Micheletti, MD (Philadelphia, Pennsylvania); Karolyn Wanat, MD (Milwaukee, Wisconsin); Amy Chen, MD (Cromwell, Connecticut); and A. Rambi Cardones, MD (Durham, North Carolina).
Pyoderma gangrenosum (PG) is a rare, chronic, ulcerative, neutrophilic dermatosis of unclear etiology. Large, multicentered, randomized controlled trials (RCTs) are challenging due to the rarity of PG and the lack of a diagnostic confirmatory test; therefore, evidence-based guidelines for diagnosis and treatment are not well established. Current management of PG primarily is guided by case series, small clinical trials, and expert opinion.1-4 We conducted a survey of expert medical dermatologists to highlight best practices in diagnostic and therapeutic approaches to PG.
Methods
The Society of Dermatology Hospitalists (SDH) Scientific Task Force gathered expert opinions from members of the SDH and Rheumatologic Dermatology Society (RDS) regarding PG workup and treatment through an online survey of 15 items (eTable 1). Subscribers of the SDH and RDS LISTSERVs were invited via email to participate in the survey from January 2016 to February 2016. Anonymous survey responses were collected and collated using SurveyMonkey. The survey results identified expert recommendations for evaluation, diagnosis, and treatment of PG and are reported as the sum of the percentage of respondents who answered always (almost 100% of the time) or often (more than half the time) following a particular course of action. A subanalysis was performed defining 2 groups of respondents based on the number of cases of PG treated per year (≥10 vs <10). Survey responses between each group were compared using χ2 analysis with statistical significance set at P=.05.
Results
Fifty-one respondents completed the survey out of 140 surveyed (36% response rate). All respondents were dermatologists, and 96% (49/51) were affiliated with an academic institution. Among the respondents, the number of PG cases managed per year ranged from 2 to 35.
Respondents consistently ordered skin biopsies (92% [47/51]) and tissue cultures (90% [46/51]), as well as certain ancillary tests, including complete blood cell count (96% [49/51]), complete metabolic panel (86% [44/51]), serum protein electrophoresis (76% [39/51]), and hepatitis panel (71% [36/51]). Other frequently ordered studies were rheumatoid factor (69% [35/51]), antinuclear antibodies (67% [34/51]), and antineutrophilic antibodies (65% [33/51]). Respondents frequently ordered erythrocyte sedimentation rate (59% [30/51]), C-reactive protein (55% [28/51]), cryoglobulins (53% [27/51]), urine protein electrophoresis (53% [27/51]), hypercoagulability workup (49% [25/51]), and serum immunofixation test (49% [25/51]). Human immunodeficiency virus testing (43% [22/51]), chest radiograph (41% [21/51]), colonoscopy (41% [21/51]) and referral to other specialties for workup—gastroenterology (38% [19/51]), hematology/oncology (14% [7/51]), and rheumatology (10% [5/51])—were less frequently ordered (eTable 2).
Systemic corticosteroids were reported as first-line therapy by most respondents (94% [48/51]), followed by topical immunomodulatory therapies (63% [32/51]). Topical corticosteroids (75% [38/51]) were the most common first-line topical agents. Thirty-nine percent of respondents (20/51) prescribed topical calcineurin inhibitors as first-line topical therapy. Additional therapies frequently used included systemic cyclosporine (47% [24/51]), antineutrophilic agents (41% [21/51]), and biologic agents (37% [19/51]). Fifty-seven percent of respondents (29/51) supported using combination topical and systemic therapy (Table).
A wide variety of wound care practices were reported in the management of PG. Seventy-six percent of respondents (39/51) favored petroleum-impregnated gauze, 69% (35/51) used nonadhesive dressings, and 43% (22/51) added antimicrobial therapy for PG wound care (eTable 3). In the subanalysis, there were no significant differences in the majority of answer responses in patients treating 10 or more PG cases per year vs fewer than 10 PG cases, except with regard to the practice of combination therapy. Those treating more than 10 cases of PG per year more frequently reported use of combination therapies compared to respondents treating fewer than 10 cases (P=.04).
Comment
Skin biopsies and tissue cultures were strongly recommended (>90% survey respondents) for the initial evaluation of lesions suspected to be PG to evaluate for typical histopathologic changes that appear early in the disease, to rule out PG mimickers such as infectious or vascular causes, and to prevent the detrimental effects of inappropriate treatment and delayed diagnosis.5
Suspected PG warrants a reasonable search for related conditions because more than 50% of PG cases are associated with comorbidities such as rheumatoid arthritis, inflammatory bowel disease, and hematologic disease/malignancy.6,7 A complete blood cell count and comprehensive metabolic panel were recommended by most respondents, aiding in the preliminary screening for hematologic and infectious causes as well as detecting liver and kidney dysfunction associated with systemic conditions. Additionally, exclusion of infection or malignancy may be particularly important if the patient will undergo systemic immunosuppression. In challenging PG cases when initial findings are inconclusive and the clinical presentation does not direct workup (eg, colonoscopy to evaluate gastrointestinal tract symptoms), serum protein electrophoresis, hepatitis panel, rheumatoid factor, antinuclear antibodies, and antineutrophilic antibody tests also were frequently ordered by respondents to further evaluate for underlying or associated conditions.
This consensus regarding skin biopsies and certain ancillary tests is consistent with the proposed diagnostic criteria for classic ulcerative PG in which the absence or exclusion of other relevant causes of cutaneous ulcers is required based on the criteria.8 The importance of ensuring an accurate diagnosis is paramount, as a 10% misdiagnosis rate has been documented in the literature.5
Importantly, a stepwise diagnostic workup for PG is proposed based on survey results, which may limit unnecessary testing and the associated costs to the health care system (Figure 1). Selection of additional testing is guided by initial test results and features of the patient’s clinical presentation, including age, review of systems, and associated comorbidities. Available data suggest that underlying inflammatory bowel disease is more frequent in PG patients who are younger than 65 years, whereas those who are 65 years and older are more likely to have inflammatory arthritis, cancer, or an underlying hematologic disorder.9
Treatment of PG should address both the inflammatory and wound components of the disease (Figure 2).7 In our survey results, systemic corticosteroids were identified as an important first-line therapy supported by reasonable evidence and were favored for their rapid response and minimal cost.1,10,11 Many respondents endorsed the use of systemic therapy in combination with topical steroids or calcineurin inhibitors. Combination therapy may provide more immediate control of rapidly progressing disease while minimizing adverse effects of long-term systemic corticosteroid use. A survey of German wound experts similarly endorsed frequent use of topical calcineurin inhibitors and combination systemic and topical glucocorticoid therapy as common therapeutic approaches.1
Importantly, treatments may vary depending on patient characteristics, comorbidities, and underlying disease, which underscores the need for individualized treatment approaches. Alternative first-line systemic treatments favored by respondents were cyclosporine, biologic medications, and antineutrophilic agents such as dapsone. Cyclosporine has demonstrated comparable efficacy to systemic glucocorticoids in one RCT and is considered an important steroid-sparing alternative for PG treatment.2 Biologic agents, especially tumor necrosis factor inhibitors, may be effective in treating cases of refractory PG or for concomitant inflammatory bowel disease management, as demonstrated by a small RCT documenting improvement of PG following infliximab infusion.3
Respondents strongly recommended petrolatum-impregnated gauze and other nonadhesive dressings, including alginate and hydrocolloid dressings, as part of PG wound care. Topical antimicrobials and compression stockings also were recommended by respondents. These practices aim to promote moist environments for healing, avoid maceration, prevent superinfection, optimize wound healing, and minimize damage from adhesive injury.12 Wound debridement and grafting generally were not recommended. However, pathergy is not a universal phenomenon in PG, and wounds that are no longer in the inflammatory phase may benefit from gentle debridement of necrotic tissue and/or grafting in select cases.10
Conclusion
An approach to modifying PG management based on clinical presentation and the practice of combination therapy with multiple systemic agents in refractory PG cases was not addressed in our survey. The low response rate is a limitation; however, the opinions of 51 medical dermatologist experts who regularly manage PG (in contrast to papers based on individualized clinical experience) can provide important clinical guidance until more scientific evidence is established.
Acknowledgments
We would like to thank the SDH and RDS membership for their participation in this survey. We especially acknowledge the other members of the SDH Scientific Task Force for their feedback: Misha Rosenbach, MD (Philadelphia, Pennsylvania); Robert G. Micheletti, MD (Philadelphia, Pennsylvania); Karolyn Wanat, MD (Milwaukee, Wisconsin); Amy Chen, MD (Cromwell, Connecticut); and A. Rambi Cardones, MD (Durham, North Carolina).
- Al Ghazal P, Dissemond J. Therapy of pyoderma gangrenosum in Germany: results of a survey among wound experts. J Dtsch Dermatol Ges . 2015;13:317-324.
- Ormerod AD, Thomas KS, Craig FE, et al. Comparison of the two most commonly used treatments for pyoderma gangrenosum: results of the STOP GAP randomised controlled trial. BMJ. 2015;350:h2958.
- Brooklyn TN, Dunnill MG, Shetty A, et al. Infliximab for the treatment of pyoderma gangrenosum: a randomised, double blind, placebo controlled trial. Gut. 2006;55:505-509.
- Al Ghazal P, Klode J, Dissemond J. Diagnostic criteria for pyoderma gangrenosum: results of a survey among dermatologic wound experts in Germany. J Dtsch Dermatol Ges. 2014;12:1129-1131.
- Weenig RH, Davis MD, Dahl PR, et al. Skin ulcers misdiagnosed as pyoderma gangrenosum. N Engl J Med. 2002;347:1412-1418.
- Powell FC, Su WP, Perry HO. Pyoderma gangrenosum: classification and management. J Am Acad Dermatol. 1996;34:395-409.
- Bennett ML, Jackson JM, Jorizzo JL, et al. Pyoderma gangrenosum: a comparison of typical and atypical forms with an emphasis on time to remission. case review of 86 patients from 2 institutions. Medicine. 2000;79:37-46.
- Su WP, Davis MD, Weening RH, et al. Pyoderma gangrenosum: clinicopathologic correlation and proposed diagnostic criteria. Int J Dermatol. 2004;43:790-800.
- Aschyan H, Butler DC, Nelson CA, et al. The association of age with clinical presentation and comorbidities of pyoderma gangrenosum. JAMA Dermatol. 2018;154:409-413.
- Binus AM, Qureshi AA, Li VW, et al. Pyoderma gangrenosum: a retrospective review of patient characteristics, comorbidities and therapy in 103 patients. Br J Dermatol. 2011;165:1244-1250.
- Reichrath J, Bens G, Bonowitz A, et al. Treatment recommendations for pyoderma gangrenosum: an evidence-based review of the literature based on more than 350 patients. J Am Acad Dermatol. 2005;53:273-283.
- Miller J, Yentzer BA, Clark A, et al. Pyoderma gangrenosum: a review and update on new therapies. J Am Acad Dermatol. 2010;62:646-654.
- Al Ghazal P, Dissemond J. Therapy of pyoderma gangrenosum in Germany: results of a survey among wound experts. J Dtsch Dermatol Ges . 2015;13:317-324.
- Ormerod AD, Thomas KS, Craig FE, et al. Comparison of the two most commonly used treatments for pyoderma gangrenosum: results of the STOP GAP randomised controlled trial. BMJ. 2015;350:h2958.
- Brooklyn TN, Dunnill MG, Shetty A, et al. Infliximab for the treatment of pyoderma gangrenosum: a randomised, double blind, placebo controlled trial. Gut. 2006;55:505-509.
- Al Ghazal P, Klode J, Dissemond J. Diagnostic criteria for pyoderma gangrenosum: results of a survey among dermatologic wound experts in Germany. J Dtsch Dermatol Ges. 2014;12:1129-1131.
- Weenig RH, Davis MD, Dahl PR, et al. Skin ulcers misdiagnosed as pyoderma gangrenosum. N Engl J Med. 2002;347:1412-1418.
- Powell FC, Su WP, Perry HO. Pyoderma gangrenosum: classification and management. J Am Acad Dermatol. 1996;34:395-409.
- Bennett ML, Jackson JM, Jorizzo JL, et al. Pyoderma gangrenosum: a comparison of typical and atypical forms with an emphasis on time to remission. case review of 86 patients from 2 institutions. Medicine. 2000;79:37-46.
- Su WP, Davis MD, Weening RH, et al. Pyoderma gangrenosum: clinicopathologic correlation and proposed diagnostic criteria. Int J Dermatol. 2004;43:790-800.
- Aschyan H, Butler DC, Nelson CA, et al. The association of age with clinical presentation and comorbidities of pyoderma gangrenosum. JAMA Dermatol. 2018;154:409-413.
- Binus AM, Qureshi AA, Li VW, et al. Pyoderma gangrenosum: a retrospective review of patient characteristics, comorbidities and therapy in 103 patients. Br J Dermatol. 2011;165:1244-1250.
- Reichrath J, Bens G, Bonowitz A, et al. Treatment recommendations for pyoderma gangrenosum: an evidence-based review of the literature based on more than 350 patients. J Am Acad Dermatol. 2005;53:273-283.
- Miller J, Yentzer BA, Clark A, et al. Pyoderma gangrenosum: a review and update on new therapies. J Am Acad Dermatol. 2010;62:646-654.
Practice Points
- The diagnosis of pyoderma gangrenosum (PG) poses a challenge in clinical practice that could be minimized by following a stepwise algorithm based on initial test results (including skin biopsies) and features of the patient’s clinical presentation.
- As there is no US Food and Drug Administration–approved treatment for PG, a stepwise algorithm approach in combination with the clinical experience addressing inflammation and wound care is essential to reach control and remission of PG.
Approximation of Alcohol-Based Hand Sanitizer Volume Using a Toothpaste Cap
Practice Gap
The Centers for Disease Control and Prevention recommends handwashing with soap and water or using alcohol-based hand sanitizers to prevent transmission of coronavirus disease 2019. Five steps are delineated for effective handwashing: wetting, lathering, scrubbing, rinsing, and drying. Although alcohol-based sanitizers may be perceived as more damaging to the skin, they are less likely to cause dermatitis than handwashing with soap and water.1 Instructions are precise for handwashing, while there are no recommendations for effective use of alcohol-based hand sanitizers. A common inquiry regarding alcohol-based hand sanitizers is the volume needed for efficacy without causing skin irritation.
The Technique
Approximately 1 mL of alcohol-based hand sanitizer is recommended by some manufacturers. However, abundant evidence refutes this recommendation, including a study that tested the microbial efficacy of alcohol-based sanitizers by volume. A volume of 2 mL was necessary to achieve the 2.0 log reduction of contaminants as required by the US Food and Drug Administration for antimicrobial efficacy.2 The precise measurement of hand sanitizer using a calibrated syringe before each use is impractical. Thus, we recommend using a screw-top toothpaste cap to assist in approximating the necessary volume (Figure). The cap holds approximately 1 mL of liquid as measured using a syringe; therefore, 2 caps filled with sanitizer should be used.
Practice Implications
The general public may be underutilizing hand sanitizer due to fear of excessive skin irritation or supply shortages, which will reduce efficacy. Patients and physicians can use this simple visual approximation to ensure adequate use of hand sanitizer volume.
- Stutz N, Becker D, Jappe U, et al. Nurses’ perceptions of the benefits and adverse effects of hand disinfection: alcohol-based hand rubs vs. hygienic handwashing: a multicentre questionnaire study with additional patch testing by the German Contact Dermatitis Research Group. Br J Dermatol. 2009;160:565-572.
- Kampf G, Ruselack S, Eggerstedt S, et al. Less and less-influence of volume on hand coverage and bactericidal efficacy in hand disinfection. BMC Infect Dis. 2013;13:472.
Practice Gap
The Centers for Disease Control and Prevention recommends handwashing with soap and water or using alcohol-based hand sanitizers to prevent transmission of coronavirus disease 2019. Five steps are delineated for effective handwashing: wetting, lathering, scrubbing, rinsing, and drying. Although alcohol-based sanitizers may be perceived as more damaging to the skin, they are less likely to cause dermatitis than handwashing with soap and water.1 Instructions are precise for handwashing, while there are no recommendations for effective use of alcohol-based hand sanitizers. A common inquiry regarding alcohol-based hand sanitizers is the volume needed for efficacy without causing skin irritation.
The Technique
Approximately 1 mL of alcohol-based hand sanitizer is recommended by some manufacturers. However, abundant evidence refutes this recommendation, including a study that tested the microbial efficacy of alcohol-based sanitizers by volume. A volume of 2 mL was necessary to achieve the 2.0 log reduction of contaminants as required by the US Food and Drug Administration for antimicrobial efficacy.2 The precise measurement of hand sanitizer using a calibrated syringe before each use is impractical. Thus, we recommend using a screw-top toothpaste cap to assist in approximating the necessary volume (Figure). The cap holds approximately 1 mL of liquid as measured using a syringe; therefore, 2 caps filled with sanitizer should be used.
Practice Implications
The general public may be underutilizing hand sanitizer due to fear of excessive skin irritation or supply shortages, which will reduce efficacy. Patients and physicians can use this simple visual approximation to ensure adequate use of hand sanitizer volume.
Practice Gap
The Centers for Disease Control and Prevention recommends handwashing with soap and water or using alcohol-based hand sanitizers to prevent transmission of coronavirus disease 2019. Five steps are delineated for effective handwashing: wetting, lathering, scrubbing, rinsing, and drying. Although alcohol-based sanitizers may be perceived as more damaging to the skin, they are less likely to cause dermatitis than handwashing with soap and water.1 Instructions are precise for handwashing, while there are no recommendations for effective use of alcohol-based hand sanitizers. A common inquiry regarding alcohol-based hand sanitizers is the volume needed for efficacy without causing skin irritation.
The Technique
Approximately 1 mL of alcohol-based hand sanitizer is recommended by some manufacturers. However, abundant evidence refutes this recommendation, including a study that tested the microbial efficacy of alcohol-based sanitizers by volume. A volume of 2 mL was necessary to achieve the 2.0 log reduction of contaminants as required by the US Food and Drug Administration for antimicrobial efficacy.2 The precise measurement of hand sanitizer using a calibrated syringe before each use is impractical. Thus, we recommend using a screw-top toothpaste cap to assist in approximating the necessary volume (Figure). The cap holds approximately 1 mL of liquid as measured using a syringe; therefore, 2 caps filled with sanitizer should be used.
Practice Implications
The general public may be underutilizing hand sanitizer due to fear of excessive skin irritation or supply shortages, which will reduce efficacy. Patients and physicians can use this simple visual approximation to ensure adequate use of hand sanitizer volume.
- Stutz N, Becker D, Jappe U, et al. Nurses’ perceptions of the benefits and adverse effects of hand disinfection: alcohol-based hand rubs vs. hygienic handwashing: a multicentre questionnaire study with additional patch testing by the German Contact Dermatitis Research Group. Br J Dermatol. 2009;160:565-572.
- Kampf G, Ruselack S, Eggerstedt S, et al. Less and less-influence of volume on hand coverage and bactericidal efficacy in hand disinfection. BMC Infect Dis. 2013;13:472.
- Stutz N, Becker D, Jappe U, et al. Nurses’ perceptions of the benefits and adverse effects of hand disinfection: alcohol-based hand rubs vs. hygienic handwashing: a multicentre questionnaire study with additional patch testing by the German Contact Dermatitis Research Group. Br J Dermatol. 2009;160:565-572.
- Kampf G, Ruselack S, Eggerstedt S, et al. Less and less-influence of volume on hand coverage and bactericidal efficacy in hand disinfection. BMC Infect Dis. 2013;13:472.
Skin Tightening
Minimally and noninvasive skin tightening has become one of the most requested cosmetic procedures. Skin laxity often is apparent in areas of the face, neck, jawline, hands, abdomen, and thighs, with features of fine lines, wrinkles, and cellulite. Intrinsic and extrinsic factors contribute to the development of skin laxity. Intrinsic aspects include chronological age, stress, and genetics, whereas extrinsic influences include exposure to solar radiation, environmental toxins, and smoking.1,2 These factors affect the production and maintenance of both collagen and elastic proteins, which are the main components that help the skin stay firm and smooth. With a goal of improving skin laxity, multiple skin tightening modalities have been developed.
Traditionally, skin laxity was treated by invasive surgical skin procedures (eg, rhytidectomy), which carry a high financial cost, require an operating room and general anesthesia, have a prolonged recovery time with notable postoperative care, and have possible risk of unwanted scars.3,4 The risks associated with invasive procedures have spurned a growing demand for minimally invasive and noninvasive methods, which have fostered the development of several skin laxity reversal modalities over the last decade. Although the achieved results of these technologies are less dramatic and require more treatments, they do not possess the associated risks and adverse effects seen in invasive surgical procedures. As such, demand for these techniques has been growing among cosmetic patients.
There are multiple technologies that currently are employed to achieve noninvasive skin tightening. Laser therapy, radiofrequency (RF), ultrasound, and intense pulsed light (IPL) are methods that focus targeted energy to elevate temperatures in the deeper layers of the skin. Elevated thermal energy causes denaturing of collagen with preservation of heat-stable intermolecular cross-links. Skin tightening is achieved through physical shortening of the collagen fibers with preservation of the heat-stable intermolecular hydrogen bonds, which leads to an increase in the rubber elastic properties of the collagen polymer and stimulation of new collagen formation.5,6 The temperature at which this process occurs has been frequently reported as approximately 65°C.7,8 Alternative noninvasive therapies that do not focus on elevated thermal energy for skin tightening include chemical peels and skin care products.
Given the multitude of treatment methods that have been developed to counteract skin laxity, this article seeks to provide an overview of some technologies, devices, and commonly used therapies to help dermatologists choose the appropriate modalities for their cosmetic patients.
Laser Therapy
Since its approval in the 1980s, laser therapy has become an alternative to invasive surgical skin tightening.9 Laser therapy utilized for treatment can be subcategorized into 2 types: ablative and nonablative.
Traditional ablative skin tightening utilized CO2 or erbium:YAG lasers. These lasers caused skin tightening by first ablating the epidermis cleanly off the dermis, with a partially coagulated area in the dermis, which triggered a wound-healing cascade followed by neocollagenesis and remodeling.10,11 Although this treatment displays notable retightening of the skin, traditional ablative lasers are not routinely used, likely because of lengthy recovery periods, risk for scar development, flares of acne and herpes simplex virus, hyperpigmentation, and delayed-onset hypopigmentation.9,12,13
Fractional ablative laser treatments soon emerged as an effective alternative to traditional ablative lasers. Various studies have noted better recovery times and side-effect profiles.14-18 This improvement is believed to be due to the method of wound healing in fractional ablative laser treatments. Ablative fractional photothermolysis works by generating deeply narrow focal ablations that involve the dermis and epidermis while leaving the surrounding skin unscathed, which allows for rapid re-epithelization, filling in of the dermal pockets, and stimulation of dermal remodeling.10,11,18,19 Studies have demonstrated a range of improvement in skin laxity from 56% to 65.3% at 6 months posttreatment.20,21 Although the incidence of reported side effects is better than with the traditional ablative laser, fractional ablative lasers have documented reports of similar types of side effects as traditional lasers due in part to ablation of the skin.22,23
Nonablative lasers were developed as alternatives to ablative laser treatments. This class of lasers produces a milder effect compared with its ablative counterpart. Studies show a quantitative improvement range of 8.9% to 11% in skin laxity 3 months posttreatment.24,25 Nonablative lasers induce controlled tissue injury in the dermis, which leads to stimulation of dermal remodeling and collagen production.11 Although the effects of nonablative lasers are milder compared with their ablative counterparts, they possess the superior benefit of minimal adverse events. Most studies reported transient erythema posttreatment, but no long-term adverse effects have been noted,26-31 in part due to preservation of the epidermal layer.
Radiofrequency
Radiofrequency technology was the first method marketed for noninvasive skin tightening. Radiofrequency devices work by generating heat through tissue resistance to an applied alternating electrical current, which leads to collagen contraction and remodeling along with neocollgenesis.32 The major electrode configurations used in these technologies are monopolar, bipolar, and multipolar, which differ by the electric field they produce. Reported side effects include erythema that arose 1 week following completion of treatment and resolved by 6-month follow-up, as well as hypertrophic scarring, transient postinflammatory hyperpigmentation, and pain.33,34
Monopolar systems were the first among these devices to be developed for use in skin tightening and remain the most extensively studied technology for treatment of skin laxity. Developed in 2001, the Thermage device (Solta Medical, Valent Pharmaceuticals) remains the most extensively studied technology for the treatment of skin laxity.35 In a trial performed by Fitzpatrick et al,36 treatment of skin laxity of the periorbital area with ThermaCool TC (Thermage, Inc) demonstrated an 83.2% improvement in at least 1 point treated and an overall 28.9% improvement of the entire treatment area at 6-month follow-up. Additionally, a survey study of 5700 patients who received monopolar RF skin tightening treatments demonstrated that 26% of patients experienced immediate tightening following treatment, and 54% observed tightening 6 months posttreatment.37
Bipolar and multipolar devices were developed following the success of monopolar devices in the treatment for skin laxity. In a study evaluating multipolar RF for the face and neck, all 11 patients were determined to have improvement of their skin laxity following weekly treatments for 8 weeks.38
Ultrasound
The use of ultrasound for skin tightening was first approved in 2009.39 The primary mechanism of skin tightening is through thermally induced contraction of collagen with subsequent collagen neogenesis achieved through absorption of the vibrational acoustic energy into target tissue.40 There are 2 types of ultrasound methods: microfocused and high-intensity focused. Microfocused ultrasound focuses on delivering lower-energy pulses to the deep reticular dermal and subdermal layers that lead to disruption of the underlying architecture of the skin, promoting increases in distensibility, elasticity, and viscoelasticity.41 To date, microfocused ultrasound is approved for treating skin laxity of the eyebrow and submental area and wrinkles of the décolleté. Currently, there are 2 devices approved by the US Food and Drug Administration for the treatment of skin laxity with ultrasound. These devices are the Ulthera System (Merz Pharmaceuticals) and the Sofwave system (Sofwave Medical Ltd).42 Oni et al43 evaluated 93 patients following treatment using Ulthera for skin laxity in the lower face. There was a noticeable improvement of 63.6% at 90 days following treatment. Brobst et al44 showed improvement in laxity at 6 months and 1.5 years following last treatment. The most commonly reported posttreatment side effects include transient purpura, transient edema, and transient postinflammatory pigmentation.42,45 Serious complications are rare and include development of palpable subcutaneous nodules and motor nerve paresis.42,46
High-intensity focused ultrasound has been more recently introduced as a modality for skin tightening and rejuvenation. This method focuses on applying heat to areas through acoustic energy to areas of the deep dermis, subdermal connective tissue, and fibromuscular layer in targeted microcoagulation zones without effect to the epidermis.47 The targeted thermal effects and microcoagulation are believed to cause skin tightening through collagen contraction and remodeling. Future studies are needed to determine the overall benefits in skin laxity to achieve approval by the US Food and Drug Administration for use as a treatment option.
IPL Therapy
Intense pulsed light therapy is different from lasers in that it utilizes a wider variety of wavelengths ranging from approximately 500 to 1200 nm.48 The process of skin tightening is achieved through selective photothermolysis in which thermal damage is focused solely on pigmented targets at the cellular or tissue levels in the epidermis and dermis.49 Intense pulsed light penetrates the tissues and is selectively absorbed by melanin and hemoglobin, thereby producing photothermal effects. The photothermal effects lead to reversible thermal damage to surrounding collagen and induction contraction of collagen fibers and fiber remodeling.50 Clinical studies on the effectiveness on skin tightening have shown incongruent results. Multiple studies have noted improvement in skin elasticity as well as increased deposits of collagen in treated areas. Other studies have shown no improvement of rhytides or wrinkle reduction. The side effects noted were transient pain, swelling, and erythema, along with rare instances of blisters and crusting.48,51-54 Due to the inhomogeneous results, the use of IPL is largely reserved for treatment of acne, hyperpigmentation, hypertrichosis, and superficial vascular malformations.
Chemical Peels
Chemical peels are used in the treatment of skin laxity through a process similar to ablative lasers. Unlike other methods described in this article, this type of treatment is only reserved for the facial areas. The peel must penetrate to the lower papillary dermis or deeper to allow for adequate collagen synthesis.55 As such, medium to deep peeling agents should be used.56 Peels cause coagulation of membrane proteins and necrosis of the epidermis and dermis, thereby stimulating collagen synthesis and keratinocyte regeneration. Additionally, there is an increase in the deposition of glycosaminoglycans, which play a major role in providing hydration for the skin because of their water-binding capacity.56 Deep peels have the added effect of restoring dermal architecture to its native state. Medium-depth peels work up to the layer of the epidermis and dermis.57 Trichloroacetic acid (TCA) 35% is the main ingredient used in these types of peels. Some examples include Monet combination (Jessner solution with 35% TCA), Brody combination (solid CO2 plus 35% TCA), and Coleman combination (70% glycolic acid and 35% TCA). Deep peels penetrate to the levels of the reticular dermis.58 The formulation of these peels contain croton oil and phenols in various concentrations.57,58 A study by Brody59 noted clinical improvement of skin laxity–attributed histologic depth achieved by medium-depth peels. The results of the study demonstrated that the depth of wounding from 3 consecutive applications of TCA led to greater epidermal hyperplasia and a more dense formation of dermal elastic fiber formation on histologic examination. Side effects noted in the study included transient erythema, edema, and erosions that resolved without scar formation at 30-day follow-up.59 Another study performed by Oresajo et al60 demonstrated that patients treated with either a chemical peel of 41% capryloyl salicylic acid or 30% glycolic acid led to notable reduction of fine lines/wrinkles vs baseline. Side effects noted included pruritus, erythema, increased skin sensitivity, epidermolysis, allergic and irritant contact dermatitis, and postinflammatory hyperpigmentation.60
Skin Care
Skin care products have been developed over the years and marketed to aid in the treatment of skin laxity. Some studied methods include photoprotection products, antioxidant-based products, and vitamin A products. Photoprotection plays a crucial role in the prevention of skin laxity. Unprotected sun exposure can induce damage to previously treated skin, leading to minimized or cancelled rejuvenation measures.61
Oxidation is a major contributor in the development of skin laxity. The skin naturally possesses endogenous antioxidant defense mechanisms that protect its cells from free radical damage. However, these mechanisms are reduced as skin ages and are further diminished with photodamage. Ascorbic acid is a collagen stimulator that is known to have antioxidant properties. In the appropriate formulations, topical vitamin C directly supplements the skin’s antioxidant reservoir.61
The use of vitamin A, a retinoic acid, for treatment of skin laxity is based on its ability to improve the production of procollagen and elastic fiber components, resulting in the restoration of dermal matrix proteins.61-65 Vitamin A in the skin plays a key role in the regulation and control of proliferation and differentiation of all major cell types found in the epidermis and dermis.61 Studies have shown that the long-term use of topical vitamin A improves fine and coarse wrinkling.65
Final Thoughts
Various technologies have been developed to provide clinically significant skin laxity reversal. Laser, RF, ultrasound, IPL, and topical therapies provide numerous options at our disposal. Although many devices are available, it is important to consider the desired outcome, cost, and adverse events when discussing therapeutic options for treating skin laxity (eTable). Patients should be advised that multiple treatment sessions over the course of months will likely be necessary. With the development of numerous technologies, we now have many options to offer our patients who desire minimally or noninvasive skin tightening.
- McCullough JL, Kelly KM. Prevention and treatment of skin aging. Ann NY Acad Sci. 2006;1067:323-331.
- Yaar M. Clinical and histological features of intrinsic versus extrinsic skin aging. In: Gilchrest BA, Krutmann J, eds. Skin Aging. Berlin, Germany: Springer, Heidelberg; 2006:9-21.
- Ramanadham SR, Costa CR, Narasimhan K, et al. Refining the anesthesia management of the face-lift patient: lessons learned from 1089 consecutive face lifts. Plast Reconstr Surg. 2015;135:723-730.
- Gupta V, Winocour J, Shi H, et al. Preoperative risk factors and complication rates in facelift: analysis of 11,300 patients. Aesthet Surg J. 2016;36:1-13.
- le Lous M, Flandin F, Herbage D, et al. Influence of collagen denaturation on the chemorheological properties of skin, assessed by differential scanning calorimetry and hydrothermal isometric tension measurement. Biochim Biophys Acta. 1982;717:295-300.
- Ross EV, Yashar SS, Naseef GS, et al. A pilot study of in vivo immediate tissue contraction with CO2 skin laser resurfacing in a live farm pig. Dermatol Surg. 1999;25:851-856.
- Arnoczky SP, Aksan A. Thermal modification of connective tissues: basic science considerations and clinical implications. J Am Acad Orthop Surg. 2000;8:305-313.
- Hsu TS, Kaminer MS. The use of nonablative radiofrequency technology to tighten the lower face and neck. Semin Cutan Med Surg. 2003;22:115-123.
- Alster TS. Cutaneous resurfacing with CO2 and erbium: YAG lasers: preoperative, intraoperative, and postoperative considerations. Plast Reconstr Surg. 1999;103:619-632; discussion 633-634.
- Omi T, Numano K. The role of the CO2 laser and fractional CO2 laser in dermatology. Laser Ther. 2014;23:49-60.
- Preissig J, Hamilton K, Markus R. Current laser resurfacing technologies: a review that delves beneath the surface. Semin Plast Surg. 2012;26:109-116.
- Bernstein LJ, Kauvar AN, Grossman MC, et al. The short‐ and long‐term side effects of carbon dioxide laser resurfacing. Dermatol Surg. 1997;23:519-525.
- Nanni CA, Alster TS. Complications of carbon dioxide laser resurfacing. an evaluation of 500 patients. Dermatol Surg. 1998;24:315-320.
- Ortiz AE, Tremaine AM, Zachary CB. Long‐term efficacy of a fractional resurfacing device. Lasers Surg Med. 2010;42:168-170.
- Rahman Z, MacFalls H, Jiang K, et al. Fractional deep dermal ablation induces tissue tightening. Lasers Surg Med. 2009;41:78-86.
- Graber EM, Tanzi EL, Alster TS. Side effects and complications of fractional laser photothermolysis: experience with 961 treatments. Dermatol Surg. 2008;34:301-305; discussion 305-307.
- Fisher GH, Geronemus RG. Short‐term side effects of fractional photothermolysis. Dermatol Surg. 2005;31:1245-1249.
- Ortiz AE, Goldman MP, Fitzpatrick RE. Ablative CO2 lasers for skin tightening: traditional versus fractional. Dermatol Surg. 2014;40(suppl 12):S147-S151.
- Geronemus RG. Fractional photothermolysis: current and future applications. Lasers Surg Med. 2006;38:169-176.
- Tierney EP, Hanke CW, Petersen J. Ablative fractionated CO2 laser treatment of photoaging: a clinical and histologic study. Dermatol Surg. 2012;38:1777-1789.
- Tierney EP, Hanke CW, Watkins L. Treatment of lower eyelid rhytids and laxity with ablative fractionated carbon-dioxide laser resurfacing: case series and review of the literature. J Am Acad Dermatol. 2011;64:730-740.
- Fife DJ, Fitzpatrick RE, Zachary CB. Complications of fractional CO2 laser resurfacing: four cases. Lasers Surg Med. 2009;41:179-184.
- Metelitsa AI, Alster TS. Fractionated laser skin resurfacing treatment complications: a review. Dermatol Surg. 2010;36:299-306.
- Miller L, Mishra V, Alsaad S, et al. Clinical evaluation of a non-ablative 1940 nm fractional laser. J Drugs Dermatol. 2014;13:1324-1329.
- Alexiades-Armenakas M. Nonablative skin tightening with a variable depth heating 1310-nm wavelength laser in combination with surface cooling. J Drugs Dermatol. 2007;6:1096-1103.
- Alster TS, Wanitphakdeedecha R. Improvement of postfractional laser erythema with light‐emitting diode photomodulation. Dermatol Surg. 2009;35:813-815.
- Fournier N, Lagarde JM, Turlier V, et al. A 35-month profilometric and clinical evaluation of non-ablative remodeling using a 1540-nm Er:glass laser. J Cosmet Laser Ther. 2004;6:126-130.
- Hædersdal M, Moreau KER, Beyer DM, et al. Fractional nonablative 1540 nm laser resurfacing for thermal burn scars: a randomized controlled trial. Lasers Surg Med. 2009;41:189-195.
- Lupton JR, Williams CM, Alster TS. Nonablative laser skin resurfacing using a 1540 nm erbium glass laser: a clinical and histologic analysis. Dermatol Surg. 2002;28:833-835.
- Moody BR, McCarthy JE, Hruza GJ. Collagen remodeling after 585‐nm pulsed dye laser irradiation: an ultrasonographic analysis. Dermatol Surg. 2003;29:997-999, discussion 999-1000.
- Pollock H, Pollock TA. NLite laser: nonablative wrinkle reduction.Aesthet Surg J. 2001;21:371-372.
- Burns JA. Thermage: monopolar radiofrequency. Aesthet Surg J. 2005;25:638-642.
- Weiss RA, Weiss MA, Munavelli G, et al. Monopolar radiofrequency facial tightening: a retrospective analysis of efficacy and safety in over 600 treatments. J Drugs Dermatol. 2006;5:707-712.
- Sadick NS, Makino Y. Selective electro‐thermolysis in aesthetic medicine: a review. Lasers Surg Med. 2004;34:91-97.
- Alster TS, Lupton JR. Nonablative cutaneous remodeling using radiofrequency devices. Clin Dermatol. 2007;25:487-491.
- Fitzpatrick R, Geronemus R, Goldberg D, et al. Multicenter study of noninvasive radiofrequency for periorbital tissue tightening. Lasers Surg Med. 2003;33:232-242.
- Dover JS, Zelickson B, 14-Physician Multispecialty Consensus Panel. Results of a survey of 5,700 patient monopolar radiofrequency facial skin tightening treatments: assessment of a low‐energy multiple‐pass technique leading to a clinical end point algorithm. Dermatol Surg. 2007;33:900-907.
- de Oliveira TC, Rocha SF, Ramos DG, et al. Effects of multipolar radiofrequency and pulsed electromagnetic field treatment for face and neck rejuvenation [published online March 8, 2017]. Dermatol Res Pract. doi:10.1155/2017/4146391.
- Fabi SG. Noninvasive skin tightening: focus on new ultrasound techniques. Clin Cosmet Investig Dermatol. 2015;8:47-52.
- Van Leenders GJ, Beerlage HP, Ruijter ET, et al. Histopathological changes associated with high intensity focused ultrasound (HIFU) treatment for localised adenocarcinoma of the prostate. J Clin Pathol. 2000;53:391-394.
- Wulkan AJ, Fabi SG, Green JB. Microfocused ultrasound for facial photorejuvenation: a review. Facial Plast Surg. 2016;32:269-275.
- MacGregor JL, Tanzi EL. Microfocused ultrasound for skin tightening. Semin Cutan Med Surg. 201332:18-25.
- Oni G, Hoxworth R, Teotia S, et al. Evaluation of a microfocused ultrasound system for improving skin laxity and tightening in the lower face. Aesthet Surg J. 2014;34:1099-1110.
- Brobst RW, Ferguson M, Perkins SW. Noninvasive treatment of the neck. Facial Plast Surg North Am. 2014;22:191-202.
- Alster TS, Tanzi EL. Noninvasive lifting of arm, thigh, and knee skin with transcutaneous intense focused ultrasound. Dermatol Surg. 2012;38:754-759.
- Missel L. Prevention of potential adverse events associated with use of Ulthera device. Tech Bull. 2011;32:18-25.
- Bove T, Zawada T, Serup J, et al. High‐frequency (20‐MHz) high‐intensity focused ultrasound (HIFU) system for dermal intervention: preclinical evaluation in skin equivalents. Skin Res Technol. 2019;25:217-228.
- Raulin C, Greve B, Grema H. IPL technology: a review. Lasers Surg Med. 2003;32:78-87.
- Anderson RR, Parrish JA. Selective photothermolysis: precise microsurgery by selective absorption of pulsed radiation. Science. 1983;220:524-527.
- Faucz LL, Will SE, Rodrigues CJ, et al. Quantitative evaluation of collagen and elastic fibers after intense pulsed light treatment of mouse skin. Lasers Surg Med. 2018;50:644-650.
- Goldberg DJ, Cutler KB. Nonablative treatment of rhytids with intense pulsed light. Lasers Surg Med. 2000;26:196-200.
- Li Y-H, Wu Y, Chen JZ, et al. Application of a new intense pulsed light device in the treatment of photoaging skin in Asian patients. Dermatol Surg. 2008;34:1459-1464.
- Shin J-W, Lee D-H, Choi S-Y, et al. Objective and non‐invasive evaluation of photorejuvenation effect with intense pulsed light treatment in Asian skin. J Eur Acad Dermatol Venereol. 2011;25:516-522.
- Weiss RA, Weiss MA, Beasley KL. Rejuvenation of photoaged skin: 5 years results with intense pulsed light of the face, neck, and chest. Dermatol Surg. 2002;28:1115-1119.
- Lee KC, Wambier CG, Soon SL, et al. Basic chemical peeling: superficial and medium-depth peels. J Am Acad Dermatol. 2019;81:313-324.
- Brody HJ. Do chemical peels tighten the skin? Dermatol Surg. 2014;40(suppl):S129-S133.
- Landau M. Chemical peels. Clin Dermatol. 2008;26:200-208.
- Meaike JD, Agrawal N, Chang D, et al. Noninvasive facial rejuvenation. part 3: physician-directed-lasers, chemical peels, and other noninvasive modalities. Semin Plast Surg. 2016;30:143-150.
- Brody HJ. Variations and comparisons in medium‐depth chemical peeling. J Dermatol Surg Oncol. 1989;15:953-963.
- Oresajo C, Yatskayer M, Hansenne I. Clinical tolerance and efficacy of capryloyl salicylic acid peel compared to a glycolic acid peel in subjects with fine lines/wrinkles and hyperpigmented skin. J Cosmet Dermatol. 2008;7:259-262.
- Aust MC, Fernandes D, Kolokythas P, et al. Percutaneous collagen induction therapy: an alternative treatment for scars, wrinkles, and skin laxity. Plast Reconstr Surg. 2008;121:1421-1429.
- Griffiths C. The role of retinoids in the prevention and repair of aged and photoaged skin. Clin Exp Dermatol. 2001;26:613-618.
- Darlenski R, Surber C, Fluhr J. Topical retinoids in the management of photodamaged skin: from theory to evidence‐based practical approach. Br J Dermatol. 2010;163:1157-1165.
- Kang S, Bergfeld W, Gottlieb AB, et al. Long-term efficacy and safety of tretinoin emollient cream 0.05% in the treatment of photodamaged facial skin: a two-year, randomized, placebo-controlled trial. Am J Clin Dermatol. 2005;6:245-253.
- Riahi RR, Bush AE, Cohen PR. Topical retinoids: therapeutic mechanisms in the treatment of photodamaged skin. Am J Clin Dermatol. 2016;17:265-276.
- American Society of Plastic Surgeons. Average surgeon/physician fees. https://www.plasticsurgery.org/documents/News/Statistics/2019/cosmetic-procedures-average-cost-2019.pdf. Accessed August 24, 2020.
Minimally and noninvasive skin tightening has become one of the most requested cosmetic procedures. Skin laxity often is apparent in areas of the face, neck, jawline, hands, abdomen, and thighs, with features of fine lines, wrinkles, and cellulite. Intrinsic and extrinsic factors contribute to the development of skin laxity. Intrinsic aspects include chronological age, stress, and genetics, whereas extrinsic influences include exposure to solar radiation, environmental toxins, and smoking.1,2 These factors affect the production and maintenance of both collagen and elastic proteins, which are the main components that help the skin stay firm and smooth. With a goal of improving skin laxity, multiple skin tightening modalities have been developed.
Traditionally, skin laxity was treated by invasive surgical skin procedures (eg, rhytidectomy), which carry a high financial cost, require an operating room and general anesthesia, have a prolonged recovery time with notable postoperative care, and have possible risk of unwanted scars.3,4 The risks associated with invasive procedures have spurned a growing demand for minimally invasive and noninvasive methods, which have fostered the development of several skin laxity reversal modalities over the last decade. Although the achieved results of these technologies are less dramatic and require more treatments, they do not possess the associated risks and adverse effects seen in invasive surgical procedures. As such, demand for these techniques has been growing among cosmetic patients.
There are multiple technologies that currently are employed to achieve noninvasive skin tightening. Laser therapy, radiofrequency (RF), ultrasound, and intense pulsed light (IPL) are methods that focus targeted energy to elevate temperatures in the deeper layers of the skin. Elevated thermal energy causes denaturing of collagen with preservation of heat-stable intermolecular cross-links. Skin tightening is achieved through physical shortening of the collagen fibers with preservation of the heat-stable intermolecular hydrogen bonds, which leads to an increase in the rubber elastic properties of the collagen polymer and stimulation of new collagen formation.5,6 The temperature at which this process occurs has been frequently reported as approximately 65°C.7,8 Alternative noninvasive therapies that do not focus on elevated thermal energy for skin tightening include chemical peels and skin care products.
Given the multitude of treatment methods that have been developed to counteract skin laxity, this article seeks to provide an overview of some technologies, devices, and commonly used therapies to help dermatologists choose the appropriate modalities for their cosmetic patients.
Laser Therapy
Since its approval in the 1980s, laser therapy has become an alternative to invasive surgical skin tightening.9 Laser therapy utilized for treatment can be subcategorized into 2 types: ablative and nonablative.
Traditional ablative skin tightening utilized CO2 or erbium:YAG lasers. These lasers caused skin tightening by first ablating the epidermis cleanly off the dermis, with a partially coagulated area in the dermis, which triggered a wound-healing cascade followed by neocollagenesis and remodeling.10,11 Although this treatment displays notable retightening of the skin, traditional ablative lasers are not routinely used, likely because of lengthy recovery periods, risk for scar development, flares of acne and herpes simplex virus, hyperpigmentation, and delayed-onset hypopigmentation.9,12,13
Fractional ablative laser treatments soon emerged as an effective alternative to traditional ablative lasers. Various studies have noted better recovery times and side-effect profiles.14-18 This improvement is believed to be due to the method of wound healing in fractional ablative laser treatments. Ablative fractional photothermolysis works by generating deeply narrow focal ablations that involve the dermis and epidermis while leaving the surrounding skin unscathed, which allows for rapid re-epithelization, filling in of the dermal pockets, and stimulation of dermal remodeling.10,11,18,19 Studies have demonstrated a range of improvement in skin laxity from 56% to 65.3% at 6 months posttreatment.20,21 Although the incidence of reported side effects is better than with the traditional ablative laser, fractional ablative lasers have documented reports of similar types of side effects as traditional lasers due in part to ablation of the skin.22,23
Nonablative lasers were developed as alternatives to ablative laser treatments. This class of lasers produces a milder effect compared with its ablative counterpart. Studies show a quantitative improvement range of 8.9% to 11% in skin laxity 3 months posttreatment.24,25 Nonablative lasers induce controlled tissue injury in the dermis, which leads to stimulation of dermal remodeling and collagen production.11 Although the effects of nonablative lasers are milder compared with their ablative counterparts, they possess the superior benefit of minimal adverse events. Most studies reported transient erythema posttreatment, but no long-term adverse effects have been noted,26-31 in part due to preservation of the epidermal layer.
Radiofrequency
Radiofrequency technology was the first method marketed for noninvasive skin tightening. Radiofrequency devices work by generating heat through tissue resistance to an applied alternating electrical current, which leads to collagen contraction and remodeling along with neocollgenesis.32 The major electrode configurations used in these technologies are monopolar, bipolar, and multipolar, which differ by the electric field they produce. Reported side effects include erythema that arose 1 week following completion of treatment and resolved by 6-month follow-up, as well as hypertrophic scarring, transient postinflammatory hyperpigmentation, and pain.33,34
Monopolar systems were the first among these devices to be developed for use in skin tightening and remain the most extensively studied technology for treatment of skin laxity. Developed in 2001, the Thermage device (Solta Medical, Valent Pharmaceuticals) remains the most extensively studied technology for the treatment of skin laxity.35 In a trial performed by Fitzpatrick et al,36 treatment of skin laxity of the periorbital area with ThermaCool TC (Thermage, Inc) demonstrated an 83.2% improvement in at least 1 point treated and an overall 28.9% improvement of the entire treatment area at 6-month follow-up. Additionally, a survey study of 5700 patients who received monopolar RF skin tightening treatments demonstrated that 26% of patients experienced immediate tightening following treatment, and 54% observed tightening 6 months posttreatment.37
Bipolar and multipolar devices were developed following the success of monopolar devices in the treatment for skin laxity. In a study evaluating multipolar RF for the face and neck, all 11 patients were determined to have improvement of their skin laxity following weekly treatments for 8 weeks.38
Ultrasound
The use of ultrasound for skin tightening was first approved in 2009.39 The primary mechanism of skin tightening is through thermally induced contraction of collagen with subsequent collagen neogenesis achieved through absorption of the vibrational acoustic energy into target tissue.40 There are 2 types of ultrasound methods: microfocused and high-intensity focused. Microfocused ultrasound focuses on delivering lower-energy pulses to the deep reticular dermal and subdermal layers that lead to disruption of the underlying architecture of the skin, promoting increases in distensibility, elasticity, and viscoelasticity.41 To date, microfocused ultrasound is approved for treating skin laxity of the eyebrow and submental area and wrinkles of the décolleté. Currently, there are 2 devices approved by the US Food and Drug Administration for the treatment of skin laxity with ultrasound. These devices are the Ulthera System (Merz Pharmaceuticals) and the Sofwave system (Sofwave Medical Ltd).42 Oni et al43 evaluated 93 patients following treatment using Ulthera for skin laxity in the lower face. There was a noticeable improvement of 63.6% at 90 days following treatment. Brobst et al44 showed improvement in laxity at 6 months and 1.5 years following last treatment. The most commonly reported posttreatment side effects include transient purpura, transient edema, and transient postinflammatory pigmentation.42,45 Serious complications are rare and include development of palpable subcutaneous nodules and motor nerve paresis.42,46
High-intensity focused ultrasound has been more recently introduced as a modality for skin tightening and rejuvenation. This method focuses on applying heat to areas through acoustic energy to areas of the deep dermis, subdermal connective tissue, and fibromuscular layer in targeted microcoagulation zones without effect to the epidermis.47 The targeted thermal effects and microcoagulation are believed to cause skin tightening through collagen contraction and remodeling. Future studies are needed to determine the overall benefits in skin laxity to achieve approval by the US Food and Drug Administration for use as a treatment option.
IPL Therapy
Intense pulsed light therapy is different from lasers in that it utilizes a wider variety of wavelengths ranging from approximately 500 to 1200 nm.48 The process of skin tightening is achieved through selective photothermolysis in which thermal damage is focused solely on pigmented targets at the cellular or tissue levels in the epidermis and dermis.49 Intense pulsed light penetrates the tissues and is selectively absorbed by melanin and hemoglobin, thereby producing photothermal effects. The photothermal effects lead to reversible thermal damage to surrounding collagen and induction contraction of collagen fibers and fiber remodeling.50 Clinical studies on the effectiveness on skin tightening have shown incongruent results. Multiple studies have noted improvement in skin elasticity as well as increased deposits of collagen in treated areas. Other studies have shown no improvement of rhytides or wrinkle reduction. The side effects noted were transient pain, swelling, and erythema, along with rare instances of blisters and crusting.48,51-54 Due to the inhomogeneous results, the use of IPL is largely reserved for treatment of acne, hyperpigmentation, hypertrichosis, and superficial vascular malformations.
Chemical Peels
Chemical peels are used in the treatment of skin laxity through a process similar to ablative lasers. Unlike other methods described in this article, this type of treatment is only reserved for the facial areas. The peel must penetrate to the lower papillary dermis or deeper to allow for adequate collagen synthesis.55 As such, medium to deep peeling agents should be used.56 Peels cause coagulation of membrane proteins and necrosis of the epidermis and dermis, thereby stimulating collagen synthesis and keratinocyte regeneration. Additionally, there is an increase in the deposition of glycosaminoglycans, which play a major role in providing hydration for the skin because of their water-binding capacity.56 Deep peels have the added effect of restoring dermal architecture to its native state. Medium-depth peels work up to the layer of the epidermis and dermis.57 Trichloroacetic acid (TCA) 35% is the main ingredient used in these types of peels. Some examples include Monet combination (Jessner solution with 35% TCA), Brody combination (solid CO2 plus 35% TCA), and Coleman combination (70% glycolic acid and 35% TCA). Deep peels penetrate to the levels of the reticular dermis.58 The formulation of these peels contain croton oil and phenols in various concentrations.57,58 A study by Brody59 noted clinical improvement of skin laxity–attributed histologic depth achieved by medium-depth peels. The results of the study demonstrated that the depth of wounding from 3 consecutive applications of TCA led to greater epidermal hyperplasia and a more dense formation of dermal elastic fiber formation on histologic examination. Side effects noted in the study included transient erythema, edema, and erosions that resolved without scar formation at 30-day follow-up.59 Another study performed by Oresajo et al60 demonstrated that patients treated with either a chemical peel of 41% capryloyl salicylic acid or 30% glycolic acid led to notable reduction of fine lines/wrinkles vs baseline. Side effects noted included pruritus, erythema, increased skin sensitivity, epidermolysis, allergic and irritant contact dermatitis, and postinflammatory hyperpigmentation.60
Skin Care
Skin care products have been developed over the years and marketed to aid in the treatment of skin laxity. Some studied methods include photoprotection products, antioxidant-based products, and vitamin A products. Photoprotection plays a crucial role in the prevention of skin laxity. Unprotected sun exposure can induce damage to previously treated skin, leading to minimized or cancelled rejuvenation measures.61
Oxidation is a major contributor in the development of skin laxity. The skin naturally possesses endogenous antioxidant defense mechanisms that protect its cells from free radical damage. However, these mechanisms are reduced as skin ages and are further diminished with photodamage. Ascorbic acid is a collagen stimulator that is known to have antioxidant properties. In the appropriate formulations, topical vitamin C directly supplements the skin’s antioxidant reservoir.61
The use of vitamin A, a retinoic acid, for treatment of skin laxity is based on its ability to improve the production of procollagen and elastic fiber components, resulting in the restoration of dermal matrix proteins.61-65 Vitamin A in the skin plays a key role in the regulation and control of proliferation and differentiation of all major cell types found in the epidermis and dermis.61 Studies have shown that the long-term use of topical vitamin A improves fine and coarse wrinkling.65
Final Thoughts
Various technologies have been developed to provide clinically significant skin laxity reversal. Laser, RF, ultrasound, IPL, and topical therapies provide numerous options at our disposal. Although many devices are available, it is important to consider the desired outcome, cost, and adverse events when discussing therapeutic options for treating skin laxity (eTable). Patients should be advised that multiple treatment sessions over the course of months will likely be necessary. With the development of numerous technologies, we now have many options to offer our patients who desire minimally or noninvasive skin tightening.
Minimally and noninvasive skin tightening has become one of the most requested cosmetic procedures. Skin laxity often is apparent in areas of the face, neck, jawline, hands, abdomen, and thighs, with features of fine lines, wrinkles, and cellulite. Intrinsic and extrinsic factors contribute to the development of skin laxity. Intrinsic aspects include chronological age, stress, and genetics, whereas extrinsic influences include exposure to solar radiation, environmental toxins, and smoking.1,2 These factors affect the production and maintenance of both collagen and elastic proteins, which are the main components that help the skin stay firm and smooth. With a goal of improving skin laxity, multiple skin tightening modalities have been developed.
Traditionally, skin laxity was treated by invasive surgical skin procedures (eg, rhytidectomy), which carry a high financial cost, require an operating room and general anesthesia, have a prolonged recovery time with notable postoperative care, and have possible risk of unwanted scars.3,4 The risks associated with invasive procedures have spurned a growing demand for minimally invasive and noninvasive methods, which have fostered the development of several skin laxity reversal modalities over the last decade. Although the achieved results of these technologies are less dramatic and require more treatments, they do not possess the associated risks and adverse effects seen in invasive surgical procedures. As such, demand for these techniques has been growing among cosmetic patients.
There are multiple technologies that currently are employed to achieve noninvasive skin tightening. Laser therapy, radiofrequency (RF), ultrasound, and intense pulsed light (IPL) are methods that focus targeted energy to elevate temperatures in the deeper layers of the skin. Elevated thermal energy causes denaturing of collagen with preservation of heat-stable intermolecular cross-links. Skin tightening is achieved through physical shortening of the collagen fibers with preservation of the heat-stable intermolecular hydrogen bonds, which leads to an increase in the rubber elastic properties of the collagen polymer and stimulation of new collagen formation.5,6 The temperature at which this process occurs has been frequently reported as approximately 65°C.7,8 Alternative noninvasive therapies that do not focus on elevated thermal energy for skin tightening include chemical peels and skin care products.
Given the multitude of treatment methods that have been developed to counteract skin laxity, this article seeks to provide an overview of some technologies, devices, and commonly used therapies to help dermatologists choose the appropriate modalities for their cosmetic patients.
Laser Therapy
Since its approval in the 1980s, laser therapy has become an alternative to invasive surgical skin tightening.9 Laser therapy utilized for treatment can be subcategorized into 2 types: ablative and nonablative.
Traditional ablative skin tightening utilized CO2 or erbium:YAG lasers. These lasers caused skin tightening by first ablating the epidermis cleanly off the dermis, with a partially coagulated area in the dermis, which triggered a wound-healing cascade followed by neocollagenesis and remodeling.10,11 Although this treatment displays notable retightening of the skin, traditional ablative lasers are not routinely used, likely because of lengthy recovery periods, risk for scar development, flares of acne and herpes simplex virus, hyperpigmentation, and delayed-onset hypopigmentation.9,12,13
Fractional ablative laser treatments soon emerged as an effective alternative to traditional ablative lasers. Various studies have noted better recovery times and side-effect profiles.14-18 This improvement is believed to be due to the method of wound healing in fractional ablative laser treatments. Ablative fractional photothermolysis works by generating deeply narrow focal ablations that involve the dermis and epidermis while leaving the surrounding skin unscathed, which allows for rapid re-epithelization, filling in of the dermal pockets, and stimulation of dermal remodeling.10,11,18,19 Studies have demonstrated a range of improvement in skin laxity from 56% to 65.3% at 6 months posttreatment.20,21 Although the incidence of reported side effects is better than with the traditional ablative laser, fractional ablative lasers have documented reports of similar types of side effects as traditional lasers due in part to ablation of the skin.22,23
Nonablative lasers were developed as alternatives to ablative laser treatments. This class of lasers produces a milder effect compared with its ablative counterpart. Studies show a quantitative improvement range of 8.9% to 11% in skin laxity 3 months posttreatment.24,25 Nonablative lasers induce controlled tissue injury in the dermis, which leads to stimulation of dermal remodeling and collagen production.11 Although the effects of nonablative lasers are milder compared with their ablative counterparts, they possess the superior benefit of minimal adverse events. Most studies reported transient erythema posttreatment, but no long-term adverse effects have been noted,26-31 in part due to preservation of the epidermal layer.
Radiofrequency
Radiofrequency technology was the first method marketed for noninvasive skin tightening. Radiofrequency devices work by generating heat through tissue resistance to an applied alternating electrical current, which leads to collagen contraction and remodeling along with neocollgenesis.32 The major electrode configurations used in these technologies are monopolar, bipolar, and multipolar, which differ by the electric field they produce. Reported side effects include erythema that arose 1 week following completion of treatment and resolved by 6-month follow-up, as well as hypertrophic scarring, transient postinflammatory hyperpigmentation, and pain.33,34
Monopolar systems were the first among these devices to be developed for use in skin tightening and remain the most extensively studied technology for treatment of skin laxity. Developed in 2001, the Thermage device (Solta Medical, Valent Pharmaceuticals) remains the most extensively studied technology for the treatment of skin laxity.35 In a trial performed by Fitzpatrick et al,36 treatment of skin laxity of the periorbital area with ThermaCool TC (Thermage, Inc) demonstrated an 83.2% improvement in at least 1 point treated and an overall 28.9% improvement of the entire treatment area at 6-month follow-up. Additionally, a survey study of 5700 patients who received monopolar RF skin tightening treatments demonstrated that 26% of patients experienced immediate tightening following treatment, and 54% observed tightening 6 months posttreatment.37
Bipolar and multipolar devices were developed following the success of monopolar devices in the treatment for skin laxity. In a study evaluating multipolar RF for the face and neck, all 11 patients were determined to have improvement of their skin laxity following weekly treatments for 8 weeks.38
Ultrasound
The use of ultrasound for skin tightening was first approved in 2009.39 The primary mechanism of skin tightening is through thermally induced contraction of collagen with subsequent collagen neogenesis achieved through absorption of the vibrational acoustic energy into target tissue.40 There are 2 types of ultrasound methods: microfocused and high-intensity focused. Microfocused ultrasound focuses on delivering lower-energy pulses to the deep reticular dermal and subdermal layers that lead to disruption of the underlying architecture of the skin, promoting increases in distensibility, elasticity, and viscoelasticity.41 To date, microfocused ultrasound is approved for treating skin laxity of the eyebrow and submental area and wrinkles of the décolleté. Currently, there are 2 devices approved by the US Food and Drug Administration for the treatment of skin laxity with ultrasound. These devices are the Ulthera System (Merz Pharmaceuticals) and the Sofwave system (Sofwave Medical Ltd).42 Oni et al43 evaluated 93 patients following treatment using Ulthera for skin laxity in the lower face. There was a noticeable improvement of 63.6% at 90 days following treatment. Brobst et al44 showed improvement in laxity at 6 months and 1.5 years following last treatment. The most commonly reported posttreatment side effects include transient purpura, transient edema, and transient postinflammatory pigmentation.42,45 Serious complications are rare and include development of palpable subcutaneous nodules and motor nerve paresis.42,46
High-intensity focused ultrasound has been more recently introduced as a modality for skin tightening and rejuvenation. This method focuses on applying heat to areas through acoustic energy to areas of the deep dermis, subdermal connective tissue, and fibromuscular layer in targeted microcoagulation zones without effect to the epidermis.47 The targeted thermal effects and microcoagulation are believed to cause skin tightening through collagen contraction and remodeling. Future studies are needed to determine the overall benefits in skin laxity to achieve approval by the US Food and Drug Administration for use as a treatment option.
IPL Therapy
Intense pulsed light therapy is different from lasers in that it utilizes a wider variety of wavelengths ranging from approximately 500 to 1200 nm.48 The process of skin tightening is achieved through selective photothermolysis in which thermal damage is focused solely on pigmented targets at the cellular or tissue levels in the epidermis and dermis.49 Intense pulsed light penetrates the tissues and is selectively absorbed by melanin and hemoglobin, thereby producing photothermal effects. The photothermal effects lead to reversible thermal damage to surrounding collagen and induction contraction of collagen fibers and fiber remodeling.50 Clinical studies on the effectiveness on skin tightening have shown incongruent results. Multiple studies have noted improvement in skin elasticity as well as increased deposits of collagen in treated areas. Other studies have shown no improvement of rhytides or wrinkle reduction. The side effects noted were transient pain, swelling, and erythema, along with rare instances of blisters and crusting.48,51-54 Due to the inhomogeneous results, the use of IPL is largely reserved for treatment of acne, hyperpigmentation, hypertrichosis, and superficial vascular malformations.
Chemical Peels
Chemical peels are used in the treatment of skin laxity through a process similar to ablative lasers. Unlike other methods described in this article, this type of treatment is only reserved for the facial areas. The peel must penetrate to the lower papillary dermis or deeper to allow for adequate collagen synthesis.55 As such, medium to deep peeling agents should be used.56 Peels cause coagulation of membrane proteins and necrosis of the epidermis and dermis, thereby stimulating collagen synthesis and keratinocyte regeneration. Additionally, there is an increase in the deposition of glycosaminoglycans, which play a major role in providing hydration for the skin because of their water-binding capacity.56 Deep peels have the added effect of restoring dermal architecture to its native state. Medium-depth peels work up to the layer of the epidermis and dermis.57 Trichloroacetic acid (TCA) 35% is the main ingredient used in these types of peels. Some examples include Monet combination (Jessner solution with 35% TCA), Brody combination (solid CO2 plus 35% TCA), and Coleman combination (70% glycolic acid and 35% TCA). Deep peels penetrate to the levels of the reticular dermis.58 The formulation of these peels contain croton oil and phenols in various concentrations.57,58 A study by Brody59 noted clinical improvement of skin laxity–attributed histologic depth achieved by medium-depth peels. The results of the study demonstrated that the depth of wounding from 3 consecutive applications of TCA led to greater epidermal hyperplasia and a more dense formation of dermal elastic fiber formation on histologic examination. Side effects noted in the study included transient erythema, edema, and erosions that resolved without scar formation at 30-day follow-up.59 Another study performed by Oresajo et al60 demonstrated that patients treated with either a chemical peel of 41% capryloyl salicylic acid or 30% glycolic acid led to notable reduction of fine lines/wrinkles vs baseline. Side effects noted included pruritus, erythema, increased skin sensitivity, epidermolysis, allergic and irritant contact dermatitis, and postinflammatory hyperpigmentation.60
Skin Care
Skin care products have been developed over the years and marketed to aid in the treatment of skin laxity. Some studied methods include photoprotection products, antioxidant-based products, and vitamin A products. Photoprotection plays a crucial role in the prevention of skin laxity. Unprotected sun exposure can induce damage to previously treated skin, leading to minimized or cancelled rejuvenation measures.61
Oxidation is a major contributor in the development of skin laxity. The skin naturally possesses endogenous antioxidant defense mechanisms that protect its cells from free radical damage. However, these mechanisms are reduced as skin ages and are further diminished with photodamage. Ascorbic acid is a collagen stimulator that is known to have antioxidant properties. In the appropriate formulations, topical vitamin C directly supplements the skin’s antioxidant reservoir.61
The use of vitamin A, a retinoic acid, for treatment of skin laxity is based on its ability to improve the production of procollagen and elastic fiber components, resulting in the restoration of dermal matrix proteins.61-65 Vitamin A in the skin plays a key role in the regulation and control of proliferation and differentiation of all major cell types found in the epidermis and dermis.61 Studies have shown that the long-term use of topical vitamin A improves fine and coarse wrinkling.65
Final Thoughts
Various technologies have been developed to provide clinically significant skin laxity reversal. Laser, RF, ultrasound, IPL, and topical therapies provide numerous options at our disposal. Although many devices are available, it is important to consider the desired outcome, cost, and adverse events when discussing therapeutic options for treating skin laxity (eTable). Patients should be advised that multiple treatment sessions over the course of months will likely be necessary. With the development of numerous technologies, we now have many options to offer our patients who desire minimally or noninvasive skin tightening.
- McCullough JL, Kelly KM. Prevention and treatment of skin aging. Ann NY Acad Sci. 2006;1067:323-331.
- Yaar M. Clinical and histological features of intrinsic versus extrinsic skin aging. In: Gilchrest BA, Krutmann J, eds. Skin Aging. Berlin, Germany: Springer, Heidelberg; 2006:9-21.
- Ramanadham SR, Costa CR, Narasimhan K, et al. Refining the anesthesia management of the face-lift patient: lessons learned from 1089 consecutive face lifts. Plast Reconstr Surg. 2015;135:723-730.
- Gupta V, Winocour J, Shi H, et al. Preoperative risk factors and complication rates in facelift: analysis of 11,300 patients. Aesthet Surg J. 2016;36:1-13.
- le Lous M, Flandin F, Herbage D, et al. Influence of collagen denaturation on the chemorheological properties of skin, assessed by differential scanning calorimetry and hydrothermal isometric tension measurement. Biochim Biophys Acta. 1982;717:295-300.
- Ross EV, Yashar SS, Naseef GS, et al. A pilot study of in vivo immediate tissue contraction with CO2 skin laser resurfacing in a live farm pig. Dermatol Surg. 1999;25:851-856.
- Arnoczky SP, Aksan A. Thermal modification of connective tissues: basic science considerations and clinical implications. J Am Acad Orthop Surg. 2000;8:305-313.
- Hsu TS, Kaminer MS. The use of nonablative radiofrequency technology to tighten the lower face and neck. Semin Cutan Med Surg. 2003;22:115-123.
- Alster TS. Cutaneous resurfacing with CO2 and erbium: YAG lasers: preoperative, intraoperative, and postoperative considerations. Plast Reconstr Surg. 1999;103:619-632; discussion 633-634.
- Omi T, Numano K. The role of the CO2 laser and fractional CO2 laser in dermatology. Laser Ther. 2014;23:49-60.
- Preissig J, Hamilton K, Markus R. Current laser resurfacing technologies: a review that delves beneath the surface. Semin Plast Surg. 2012;26:109-116.
- Bernstein LJ, Kauvar AN, Grossman MC, et al. The short‐ and long‐term side effects of carbon dioxide laser resurfacing. Dermatol Surg. 1997;23:519-525.
- Nanni CA, Alster TS. Complications of carbon dioxide laser resurfacing. an evaluation of 500 patients. Dermatol Surg. 1998;24:315-320.
- Ortiz AE, Tremaine AM, Zachary CB. Long‐term efficacy of a fractional resurfacing device. Lasers Surg Med. 2010;42:168-170.
- Rahman Z, MacFalls H, Jiang K, et al. Fractional deep dermal ablation induces tissue tightening. Lasers Surg Med. 2009;41:78-86.
- Graber EM, Tanzi EL, Alster TS. Side effects and complications of fractional laser photothermolysis: experience with 961 treatments. Dermatol Surg. 2008;34:301-305; discussion 305-307.
- Fisher GH, Geronemus RG. Short‐term side effects of fractional photothermolysis. Dermatol Surg. 2005;31:1245-1249.
- Ortiz AE, Goldman MP, Fitzpatrick RE. Ablative CO2 lasers for skin tightening: traditional versus fractional. Dermatol Surg. 2014;40(suppl 12):S147-S151.
- Geronemus RG. Fractional photothermolysis: current and future applications. Lasers Surg Med. 2006;38:169-176.
- Tierney EP, Hanke CW, Petersen J. Ablative fractionated CO2 laser treatment of photoaging: a clinical and histologic study. Dermatol Surg. 2012;38:1777-1789.
- Tierney EP, Hanke CW, Watkins L. Treatment of lower eyelid rhytids and laxity with ablative fractionated carbon-dioxide laser resurfacing: case series and review of the literature. J Am Acad Dermatol. 2011;64:730-740.
- Fife DJ, Fitzpatrick RE, Zachary CB. Complications of fractional CO2 laser resurfacing: four cases. Lasers Surg Med. 2009;41:179-184.
- Metelitsa AI, Alster TS. Fractionated laser skin resurfacing treatment complications: a review. Dermatol Surg. 2010;36:299-306.
- Miller L, Mishra V, Alsaad S, et al. Clinical evaluation of a non-ablative 1940 nm fractional laser. J Drugs Dermatol. 2014;13:1324-1329.
- Alexiades-Armenakas M. Nonablative skin tightening with a variable depth heating 1310-nm wavelength laser in combination with surface cooling. J Drugs Dermatol. 2007;6:1096-1103.
- Alster TS, Wanitphakdeedecha R. Improvement of postfractional laser erythema with light‐emitting diode photomodulation. Dermatol Surg. 2009;35:813-815.
- Fournier N, Lagarde JM, Turlier V, et al. A 35-month profilometric and clinical evaluation of non-ablative remodeling using a 1540-nm Er:glass laser. J Cosmet Laser Ther. 2004;6:126-130.
- Hædersdal M, Moreau KER, Beyer DM, et al. Fractional nonablative 1540 nm laser resurfacing for thermal burn scars: a randomized controlled trial. Lasers Surg Med. 2009;41:189-195.
- Lupton JR, Williams CM, Alster TS. Nonablative laser skin resurfacing using a 1540 nm erbium glass laser: a clinical and histologic analysis. Dermatol Surg. 2002;28:833-835.
- Moody BR, McCarthy JE, Hruza GJ. Collagen remodeling after 585‐nm pulsed dye laser irradiation: an ultrasonographic analysis. Dermatol Surg. 2003;29:997-999, discussion 999-1000.
- Pollock H, Pollock TA. NLite laser: nonablative wrinkle reduction.Aesthet Surg J. 2001;21:371-372.
- Burns JA. Thermage: monopolar radiofrequency. Aesthet Surg J. 2005;25:638-642.
- Weiss RA, Weiss MA, Munavelli G, et al. Monopolar radiofrequency facial tightening: a retrospective analysis of efficacy and safety in over 600 treatments. J Drugs Dermatol. 2006;5:707-712.
- Sadick NS, Makino Y. Selective electro‐thermolysis in aesthetic medicine: a review. Lasers Surg Med. 2004;34:91-97.
- Alster TS, Lupton JR. Nonablative cutaneous remodeling using radiofrequency devices. Clin Dermatol. 2007;25:487-491.
- Fitzpatrick R, Geronemus R, Goldberg D, et al. Multicenter study of noninvasive radiofrequency for periorbital tissue tightening. Lasers Surg Med. 2003;33:232-242.
- Dover JS, Zelickson B, 14-Physician Multispecialty Consensus Panel. Results of a survey of 5,700 patient monopolar radiofrequency facial skin tightening treatments: assessment of a low‐energy multiple‐pass technique leading to a clinical end point algorithm. Dermatol Surg. 2007;33:900-907.
- de Oliveira TC, Rocha SF, Ramos DG, et al. Effects of multipolar radiofrequency and pulsed electromagnetic field treatment for face and neck rejuvenation [published online March 8, 2017]. Dermatol Res Pract. doi:10.1155/2017/4146391.
- Fabi SG. Noninvasive skin tightening: focus on new ultrasound techniques. Clin Cosmet Investig Dermatol. 2015;8:47-52.
- Van Leenders GJ, Beerlage HP, Ruijter ET, et al. Histopathological changes associated with high intensity focused ultrasound (HIFU) treatment for localised adenocarcinoma of the prostate. J Clin Pathol. 2000;53:391-394.
- Wulkan AJ, Fabi SG, Green JB. Microfocused ultrasound for facial photorejuvenation: a review. Facial Plast Surg. 2016;32:269-275.
- MacGregor JL, Tanzi EL. Microfocused ultrasound for skin tightening. Semin Cutan Med Surg. 201332:18-25.
- Oni G, Hoxworth R, Teotia S, et al. Evaluation of a microfocused ultrasound system for improving skin laxity and tightening in the lower face. Aesthet Surg J. 2014;34:1099-1110.
- Brobst RW, Ferguson M, Perkins SW. Noninvasive treatment of the neck. Facial Plast Surg North Am. 2014;22:191-202.
- Alster TS, Tanzi EL. Noninvasive lifting of arm, thigh, and knee skin with transcutaneous intense focused ultrasound. Dermatol Surg. 2012;38:754-759.
- Missel L. Prevention of potential adverse events associated with use of Ulthera device. Tech Bull. 2011;32:18-25.
- Bove T, Zawada T, Serup J, et al. High‐frequency (20‐MHz) high‐intensity focused ultrasound (HIFU) system for dermal intervention: preclinical evaluation in skin equivalents. Skin Res Technol. 2019;25:217-228.
- Raulin C, Greve B, Grema H. IPL technology: a review. Lasers Surg Med. 2003;32:78-87.
- Anderson RR, Parrish JA. Selective photothermolysis: precise microsurgery by selective absorption of pulsed radiation. Science. 1983;220:524-527.
- Faucz LL, Will SE, Rodrigues CJ, et al. Quantitative evaluation of collagen and elastic fibers after intense pulsed light treatment of mouse skin. Lasers Surg Med. 2018;50:644-650.
- Goldberg DJ, Cutler KB. Nonablative treatment of rhytids with intense pulsed light. Lasers Surg Med. 2000;26:196-200.
- Li Y-H, Wu Y, Chen JZ, et al. Application of a new intense pulsed light device in the treatment of photoaging skin in Asian patients. Dermatol Surg. 2008;34:1459-1464.
- Shin J-W, Lee D-H, Choi S-Y, et al. Objective and non‐invasive evaluation of photorejuvenation effect with intense pulsed light treatment in Asian skin. J Eur Acad Dermatol Venereol. 2011;25:516-522.
- Weiss RA, Weiss MA, Beasley KL. Rejuvenation of photoaged skin: 5 years results with intense pulsed light of the face, neck, and chest. Dermatol Surg. 2002;28:1115-1119.
- Lee KC, Wambier CG, Soon SL, et al. Basic chemical peeling: superficial and medium-depth peels. J Am Acad Dermatol. 2019;81:313-324.
- Brody HJ. Do chemical peels tighten the skin? Dermatol Surg. 2014;40(suppl):S129-S133.
- Landau M. Chemical peels. Clin Dermatol. 2008;26:200-208.
- Meaike JD, Agrawal N, Chang D, et al. Noninvasive facial rejuvenation. part 3: physician-directed-lasers, chemical peels, and other noninvasive modalities. Semin Plast Surg. 2016;30:143-150.
- Brody HJ. Variations and comparisons in medium‐depth chemical peeling. J Dermatol Surg Oncol. 1989;15:953-963.
- Oresajo C, Yatskayer M, Hansenne I. Clinical tolerance and efficacy of capryloyl salicylic acid peel compared to a glycolic acid peel in subjects with fine lines/wrinkles and hyperpigmented skin. J Cosmet Dermatol. 2008;7:259-262.
- Aust MC, Fernandes D, Kolokythas P, et al. Percutaneous collagen induction therapy: an alternative treatment for scars, wrinkles, and skin laxity. Plast Reconstr Surg. 2008;121:1421-1429.
- Griffiths C. The role of retinoids in the prevention and repair of aged and photoaged skin. Clin Exp Dermatol. 2001;26:613-618.
- Darlenski R, Surber C, Fluhr J. Topical retinoids in the management of photodamaged skin: from theory to evidence‐based practical approach. Br J Dermatol. 2010;163:1157-1165.
- Kang S, Bergfeld W, Gottlieb AB, et al. Long-term efficacy and safety of tretinoin emollient cream 0.05% in the treatment of photodamaged facial skin: a two-year, randomized, placebo-controlled trial. Am J Clin Dermatol. 2005;6:245-253.
- Riahi RR, Bush AE, Cohen PR. Topical retinoids: therapeutic mechanisms in the treatment of photodamaged skin. Am J Clin Dermatol. 2016;17:265-276.
- American Society of Plastic Surgeons. Average surgeon/physician fees. https://www.plasticsurgery.org/documents/News/Statistics/2019/cosmetic-procedures-average-cost-2019.pdf. Accessed August 24, 2020.
- McCullough JL, Kelly KM. Prevention and treatment of skin aging. Ann NY Acad Sci. 2006;1067:323-331.
- Yaar M. Clinical and histological features of intrinsic versus extrinsic skin aging. In: Gilchrest BA, Krutmann J, eds. Skin Aging. Berlin, Germany: Springer, Heidelberg; 2006:9-21.
- Ramanadham SR, Costa CR, Narasimhan K, et al. Refining the anesthesia management of the face-lift patient: lessons learned from 1089 consecutive face lifts. Plast Reconstr Surg. 2015;135:723-730.
- Gupta V, Winocour J, Shi H, et al. Preoperative risk factors and complication rates in facelift: analysis of 11,300 patients. Aesthet Surg J. 2016;36:1-13.
- le Lous M, Flandin F, Herbage D, et al. Influence of collagen denaturation on the chemorheological properties of skin, assessed by differential scanning calorimetry and hydrothermal isometric tension measurement. Biochim Biophys Acta. 1982;717:295-300.
- Ross EV, Yashar SS, Naseef GS, et al. A pilot study of in vivo immediate tissue contraction with CO2 skin laser resurfacing in a live farm pig. Dermatol Surg. 1999;25:851-856.
- Arnoczky SP, Aksan A. Thermal modification of connective tissues: basic science considerations and clinical implications. J Am Acad Orthop Surg. 2000;8:305-313.
- Hsu TS, Kaminer MS. The use of nonablative radiofrequency technology to tighten the lower face and neck. Semin Cutan Med Surg. 2003;22:115-123.
- Alster TS. Cutaneous resurfacing with CO2 and erbium: YAG lasers: preoperative, intraoperative, and postoperative considerations. Plast Reconstr Surg. 1999;103:619-632; discussion 633-634.
- Omi T, Numano K. The role of the CO2 laser and fractional CO2 laser in dermatology. Laser Ther. 2014;23:49-60.
- Preissig J, Hamilton K, Markus R. Current laser resurfacing technologies: a review that delves beneath the surface. Semin Plast Surg. 2012;26:109-116.
- Bernstein LJ, Kauvar AN, Grossman MC, et al. The short‐ and long‐term side effects of carbon dioxide laser resurfacing. Dermatol Surg. 1997;23:519-525.
- Nanni CA, Alster TS. Complications of carbon dioxide laser resurfacing. an evaluation of 500 patients. Dermatol Surg. 1998;24:315-320.
- Ortiz AE, Tremaine AM, Zachary CB. Long‐term efficacy of a fractional resurfacing device. Lasers Surg Med. 2010;42:168-170.
- Rahman Z, MacFalls H, Jiang K, et al. Fractional deep dermal ablation induces tissue tightening. Lasers Surg Med. 2009;41:78-86.
- Graber EM, Tanzi EL, Alster TS. Side effects and complications of fractional laser photothermolysis: experience with 961 treatments. Dermatol Surg. 2008;34:301-305; discussion 305-307.
- Fisher GH, Geronemus RG. Short‐term side effects of fractional photothermolysis. Dermatol Surg. 2005;31:1245-1249.
- Ortiz AE, Goldman MP, Fitzpatrick RE. Ablative CO2 lasers for skin tightening: traditional versus fractional. Dermatol Surg. 2014;40(suppl 12):S147-S151.
- Geronemus RG. Fractional photothermolysis: current and future applications. Lasers Surg Med. 2006;38:169-176.
- Tierney EP, Hanke CW, Petersen J. Ablative fractionated CO2 laser treatment of photoaging: a clinical and histologic study. Dermatol Surg. 2012;38:1777-1789.
- Tierney EP, Hanke CW, Watkins L. Treatment of lower eyelid rhytids and laxity with ablative fractionated carbon-dioxide laser resurfacing: case series and review of the literature. J Am Acad Dermatol. 2011;64:730-740.
- Fife DJ, Fitzpatrick RE, Zachary CB. Complications of fractional CO2 laser resurfacing: four cases. Lasers Surg Med. 2009;41:179-184.
- Metelitsa AI, Alster TS. Fractionated laser skin resurfacing treatment complications: a review. Dermatol Surg. 2010;36:299-306.
- Miller L, Mishra V, Alsaad S, et al. Clinical evaluation of a non-ablative 1940 nm fractional laser. J Drugs Dermatol. 2014;13:1324-1329.
- Alexiades-Armenakas M. Nonablative skin tightening with a variable depth heating 1310-nm wavelength laser in combination with surface cooling. J Drugs Dermatol. 2007;6:1096-1103.
- Alster TS, Wanitphakdeedecha R. Improvement of postfractional laser erythema with light‐emitting diode photomodulation. Dermatol Surg. 2009;35:813-815.
- Fournier N, Lagarde JM, Turlier V, et al. A 35-month profilometric and clinical evaluation of non-ablative remodeling using a 1540-nm Er:glass laser. J Cosmet Laser Ther. 2004;6:126-130.
- Hædersdal M, Moreau KER, Beyer DM, et al. Fractional nonablative 1540 nm laser resurfacing for thermal burn scars: a randomized controlled trial. Lasers Surg Med. 2009;41:189-195.
- Lupton JR, Williams CM, Alster TS. Nonablative laser skin resurfacing using a 1540 nm erbium glass laser: a clinical and histologic analysis. Dermatol Surg. 2002;28:833-835.
- Moody BR, McCarthy JE, Hruza GJ. Collagen remodeling after 585‐nm pulsed dye laser irradiation: an ultrasonographic analysis. Dermatol Surg. 2003;29:997-999, discussion 999-1000.
- Pollock H, Pollock TA. NLite laser: nonablative wrinkle reduction.Aesthet Surg J. 2001;21:371-372.
- Burns JA. Thermage: monopolar radiofrequency. Aesthet Surg J. 2005;25:638-642.
- Weiss RA, Weiss MA, Munavelli G, et al. Monopolar radiofrequency facial tightening: a retrospective analysis of efficacy and safety in over 600 treatments. J Drugs Dermatol. 2006;5:707-712.
- Sadick NS, Makino Y. Selective electro‐thermolysis in aesthetic medicine: a review. Lasers Surg Med. 2004;34:91-97.
- Alster TS, Lupton JR. Nonablative cutaneous remodeling using radiofrequency devices. Clin Dermatol. 2007;25:487-491.
- Fitzpatrick R, Geronemus R, Goldberg D, et al. Multicenter study of noninvasive radiofrequency for periorbital tissue tightening. Lasers Surg Med. 2003;33:232-242.
- Dover JS, Zelickson B, 14-Physician Multispecialty Consensus Panel. Results of a survey of 5,700 patient monopolar radiofrequency facial skin tightening treatments: assessment of a low‐energy multiple‐pass technique leading to a clinical end point algorithm. Dermatol Surg. 2007;33:900-907.
- de Oliveira TC, Rocha SF, Ramos DG, et al. Effects of multipolar radiofrequency and pulsed electromagnetic field treatment for face and neck rejuvenation [published online March 8, 2017]. Dermatol Res Pract. doi:10.1155/2017/4146391.
- Fabi SG. Noninvasive skin tightening: focus on new ultrasound techniques. Clin Cosmet Investig Dermatol. 2015;8:47-52.
- Van Leenders GJ, Beerlage HP, Ruijter ET, et al. Histopathological changes associated with high intensity focused ultrasound (HIFU) treatment for localised adenocarcinoma of the prostate. J Clin Pathol. 2000;53:391-394.
- Wulkan AJ, Fabi SG, Green JB. Microfocused ultrasound for facial photorejuvenation: a review. Facial Plast Surg. 2016;32:269-275.
- MacGregor JL, Tanzi EL. Microfocused ultrasound for skin tightening. Semin Cutan Med Surg. 201332:18-25.
- Oni G, Hoxworth R, Teotia S, et al. Evaluation of a microfocused ultrasound system for improving skin laxity and tightening in the lower face. Aesthet Surg J. 2014;34:1099-1110.
- Brobst RW, Ferguson M, Perkins SW. Noninvasive treatment of the neck. Facial Plast Surg North Am. 2014;22:191-202.
- Alster TS, Tanzi EL. Noninvasive lifting of arm, thigh, and knee skin with transcutaneous intense focused ultrasound. Dermatol Surg. 2012;38:754-759.
- Missel L. Prevention of potential adverse events associated with use of Ulthera device. Tech Bull. 2011;32:18-25.
- Bove T, Zawada T, Serup J, et al. High‐frequency (20‐MHz) high‐intensity focused ultrasound (HIFU) system for dermal intervention: preclinical evaluation in skin equivalents. Skin Res Technol. 2019;25:217-228.
- Raulin C, Greve B, Grema H. IPL technology: a review. Lasers Surg Med. 2003;32:78-87.
- Anderson RR, Parrish JA. Selective photothermolysis: precise microsurgery by selective absorption of pulsed radiation. Science. 1983;220:524-527.
- Faucz LL, Will SE, Rodrigues CJ, et al. Quantitative evaluation of collagen and elastic fibers after intense pulsed light treatment of mouse skin. Lasers Surg Med. 2018;50:644-650.
- Goldberg DJ, Cutler KB. Nonablative treatment of rhytids with intense pulsed light. Lasers Surg Med. 2000;26:196-200.
- Li Y-H, Wu Y, Chen JZ, et al. Application of a new intense pulsed light device in the treatment of photoaging skin in Asian patients. Dermatol Surg. 2008;34:1459-1464.
- Shin J-W, Lee D-H, Choi S-Y, et al. Objective and non‐invasive evaluation of photorejuvenation effect with intense pulsed light treatment in Asian skin. J Eur Acad Dermatol Venereol. 2011;25:516-522.
- Weiss RA, Weiss MA, Beasley KL. Rejuvenation of photoaged skin: 5 years results with intense pulsed light of the face, neck, and chest. Dermatol Surg. 2002;28:1115-1119.
- Lee KC, Wambier CG, Soon SL, et al. Basic chemical peeling: superficial and medium-depth peels. J Am Acad Dermatol. 2019;81:313-324.
- Brody HJ. Do chemical peels tighten the skin? Dermatol Surg. 2014;40(suppl):S129-S133.
- Landau M. Chemical peels. Clin Dermatol. 2008;26:200-208.
- Meaike JD, Agrawal N, Chang D, et al. Noninvasive facial rejuvenation. part 3: physician-directed-lasers, chemical peels, and other noninvasive modalities. Semin Plast Surg. 2016;30:143-150.
- Brody HJ. Variations and comparisons in medium‐depth chemical peeling. J Dermatol Surg Oncol. 1989;15:953-963.
- Oresajo C, Yatskayer M, Hansenne I. Clinical tolerance and efficacy of capryloyl salicylic acid peel compared to a glycolic acid peel in subjects with fine lines/wrinkles and hyperpigmented skin. J Cosmet Dermatol. 2008;7:259-262.
- Aust MC, Fernandes D, Kolokythas P, et al. Percutaneous collagen induction therapy: an alternative treatment for scars, wrinkles, and skin laxity. Plast Reconstr Surg. 2008;121:1421-1429.
- Griffiths C. The role of retinoids in the prevention and repair of aged and photoaged skin. Clin Exp Dermatol. 2001;26:613-618.
- Darlenski R, Surber C, Fluhr J. Topical retinoids in the management of photodamaged skin: from theory to evidence‐based practical approach. Br J Dermatol. 2010;163:1157-1165.
- Kang S, Bergfeld W, Gottlieb AB, et al. Long-term efficacy and safety of tretinoin emollient cream 0.05% in the treatment of photodamaged facial skin: a two-year, randomized, placebo-controlled trial. Am J Clin Dermatol. 2005;6:245-253.
- Riahi RR, Bush AE, Cohen PR. Topical retinoids: therapeutic mechanisms in the treatment of photodamaged skin. Am J Clin Dermatol. 2016;17:265-276.
- American Society of Plastic Surgeons. Average surgeon/physician fees. https://www.plasticsurgery.org/documents/News/Statistics/2019/cosmetic-procedures-average-cost-2019.pdf. Accessed August 24, 2020.
Practice Points
- There are a multitude of noninvasive modalities available to treat skin laxity.
- Understanding the mechanisms of each modality is crucial to selecting the appropriate treatment for your patients.
- Treatments should be tailored to the individual patient based on desired outcome, possible adverse events, patient preferences, and cost.
Filling Gaps: Moving Toward Better Treatment of Children With Atopic Dermatitis
It is a brand-new day for the treatment of children with severe inflammatory skin diseases. Not coincidentally, it also is a new day for the treatment of atopic dermatitis (AD). Why?
Historically, children have largely been ignored by pharmaceutical companies and the US Food and Drug Administration (FDA). Drug trials of new medications have been the exclusive province of adults; therefore, information they have generated has had only derivative relevance to the pediatric population. Pediatricians and providers who care for children, aware that they are not simply “little adults,” have been forced to extrapolate best practices.
My institution is poised to enroll a 3-year-old child with severe AD into a biologic trial (ClinicalTrials.gov identifier NCT03346434). The age range for this study is 6 months to 6 years. This extraordinary democratization of clinical trials is no accident. The Best Pharmaceuticals for Children Act, which was passed in 2002, was a first step. This legislation incentivized pharmaceutical companies to include children, who are notoriously more costly to study for myriad reasons, by extending patent protection for approved medications. Subsequent efforts spearheaded by advocacy groups such as the National Eczema Association included the production of guidance documents for industry1 and presentations directly to the FDA’s Dermatologic and Ophthalmic Drugs Advisory Committee meeting punctuated by powerful patient testimonials.2
Serendipitously, AD, a disease that presents by kindergarten in up to 90% of affected individuals, also has caught the eye of the pharmaceutical industry. Remarkable advances in the understanding of AD inflammation have led to an explosion of new therapeutic targets of interest. By way of context, between the introduction of topical calcineurin inhibitors in 2001 and the FDA approval of dupilumab and crisaborole in 2017, there were precisely zero new molecules approved for the treatment of AD. Viewed through another lens, prior to 2017, the only FDA-approved systemic medication for AD was prednisone, a drug most AD experts would list as the least appropriate choice for treatment of this condition.
Fast-forward to 2020 and we have a plethora of new possibilities. The National Eczema Association’s research web page
This confluence of better science, powerful advocacy, and enlightened self-interest has been revolutionary. It is most evident when parents/guardians—many of whom had long ago given up on new therapies for themselves—are gobsmacked by the new therapeutic landscape outlined for their children. Parents/guardians realize their children need not struggle as they may have themselves. The impact on quality of life has long been known, but several recent publications have brought it into finer relief. Drucker et al5 highlighted the overall burden of disease, and several subsequent papers have focused specifically on affective impacts including increased risk for depression, suicidal ideation, and suicide.6,7 In this issue of Cutis, Tracy et al8 provide an update on pediatric AD with an emphasis on comorbidities, quality of life, and evolving practices and therapies.
Better science, better drugs, better advocacy, better outcomes—it has not been a straight line, but it has indisputably been a forward-marching one. It is a new day, indeed.
- Siegfried EC, Jaworski JC, Eichenfield LF, et al. Developing drugs for the treatment of atopic dermatitis in children (≥3 months to <18 years of age): draft guidance for industry [published online March 30, 2018]. Pediatr Dermatol. 2018; May 35:303-322.
- Pediatric trials for AD systemic treatments. Dermatology Times. May 21, 2015. https://www.dermatologytimes.com/view/pediatric-trials-ad-systemic-treatments. Accessed August 11, 2020.
- Solimani F, Meier K, Ghoreschi K. Emerging topical and systemic JAK inhibitors in dermatology. Front Immunol. 2019;10:2847.
- Chan S, Cornelius V, Cro S, et al. Treatment effect of omalizumab on severe pediatric atopic dermatitis: the ADAPT randomized controlled trial. JAMA Pediatr. 2019;174:29-37.
- Drucker AM, Wang AR, Li W-Q, et al. The burden of atopic dermatitis: summary of a report for the National Eczema Association [published online September 8, 2016]. J Invest Dermatol. 2017;137:26-30.
- Sandhu JK, Wu KK, Bui T-L, et al. Association between atopic dermatitis and suicidality: a systematic review and meta-analysis. JAMA Dermatol. 2019;155:178-187.
- Patel KR, Immaneni S, Singam V, et al. Association between atopic dermatitis, depression, and suicidal ideation: a systematic review and meta-analysis [published online October 23, 2018]. J Am Acad Dermatol. 2019;80:402-410.
- Tracy A, Bhatti S, Eichenfield LF. Update on pediatric atopic dermatitis. Cutis. 2020;106:143-146.
It is a brand-new day for the treatment of children with severe inflammatory skin diseases. Not coincidentally, it also is a new day for the treatment of atopic dermatitis (AD). Why?
Historically, children have largely been ignored by pharmaceutical companies and the US Food and Drug Administration (FDA). Drug trials of new medications have been the exclusive province of adults; therefore, information they have generated has had only derivative relevance to the pediatric population. Pediatricians and providers who care for children, aware that they are not simply “little adults,” have been forced to extrapolate best practices.
My institution is poised to enroll a 3-year-old child with severe AD into a biologic trial (ClinicalTrials.gov identifier NCT03346434). The age range for this study is 6 months to 6 years. This extraordinary democratization of clinical trials is no accident. The Best Pharmaceuticals for Children Act, which was passed in 2002, was a first step. This legislation incentivized pharmaceutical companies to include children, who are notoriously more costly to study for myriad reasons, by extending patent protection for approved medications. Subsequent efforts spearheaded by advocacy groups such as the National Eczema Association included the production of guidance documents for industry1 and presentations directly to the FDA’s Dermatologic and Ophthalmic Drugs Advisory Committee meeting punctuated by powerful patient testimonials.2
Serendipitously, AD, a disease that presents by kindergarten in up to 90% of affected individuals, also has caught the eye of the pharmaceutical industry. Remarkable advances in the understanding of AD inflammation have led to an explosion of new therapeutic targets of interest. By way of context, between the introduction of topical calcineurin inhibitors in 2001 and the FDA approval of dupilumab and crisaborole in 2017, there were precisely zero new molecules approved for the treatment of AD. Viewed through another lens, prior to 2017, the only FDA-approved systemic medication for AD was prednisone, a drug most AD experts would list as the least appropriate choice for treatment of this condition.
Fast-forward to 2020 and we have a plethora of new possibilities. The National Eczema Association’s research web page
This confluence of better science, powerful advocacy, and enlightened self-interest has been revolutionary. It is most evident when parents/guardians—many of whom had long ago given up on new therapies for themselves—are gobsmacked by the new therapeutic landscape outlined for their children. Parents/guardians realize their children need not struggle as they may have themselves. The impact on quality of life has long been known, but several recent publications have brought it into finer relief. Drucker et al5 highlighted the overall burden of disease, and several subsequent papers have focused specifically on affective impacts including increased risk for depression, suicidal ideation, and suicide.6,7 In this issue of Cutis, Tracy et al8 provide an update on pediatric AD with an emphasis on comorbidities, quality of life, and evolving practices and therapies.
Better science, better drugs, better advocacy, better outcomes—it has not been a straight line, but it has indisputably been a forward-marching one. It is a new day, indeed.
It is a brand-new day for the treatment of children with severe inflammatory skin diseases. Not coincidentally, it also is a new day for the treatment of atopic dermatitis (AD). Why?
Historically, children have largely been ignored by pharmaceutical companies and the US Food and Drug Administration (FDA). Drug trials of new medications have been the exclusive province of adults; therefore, information they have generated has had only derivative relevance to the pediatric population. Pediatricians and providers who care for children, aware that they are not simply “little adults,” have been forced to extrapolate best practices.
My institution is poised to enroll a 3-year-old child with severe AD into a biologic trial (ClinicalTrials.gov identifier NCT03346434). The age range for this study is 6 months to 6 years. This extraordinary democratization of clinical trials is no accident. The Best Pharmaceuticals for Children Act, which was passed in 2002, was a first step. This legislation incentivized pharmaceutical companies to include children, who are notoriously more costly to study for myriad reasons, by extending patent protection for approved medications. Subsequent efforts spearheaded by advocacy groups such as the National Eczema Association included the production of guidance documents for industry1 and presentations directly to the FDA’s Dermatologic and Ophthalmic Drugs Advisory Committee meeting punctuated by powerful patient testimonials.2
Serendipitously, AD, a disease that presents by kindergarten in up to 90% of affected individuals, also has caught the eye of the pharmaceutical industry. Remarkable advances in the understanding of AD inflammation have led to an explosion of new therapeutic targets of interest. By way of context, between the introduction of topical calcineurin inhibitors in 2001 and the FDA approval of dupilumab and crisaborole in 2017, there were precisely zero new molecules approved for the treatment of AD. Viewed through another lens, prior to 2017, the only FDA-approved systemic medication for AD was prednisone, a drug most AD experts would list as the least appropriate choice for treatment of this condition.
Fast-forward to 2020 and we have a plethora of new possibilities. The National Eczema Association’s research web page
This confluence of better science, powerful advocacy, and enlightened self-interest has been revolutionary. It is most evident when parents/guardians—many of whom had long ago given up on new therapies for themselves—are gobsmacked by the new therapeutic landscape outlined for their children. Parents/guardians realize their children need not struggle as they may have themselves. The impact on quality of life has long been known, but several recent publications have brought it into finer relief. Drucker et al5 highlighted the overall burden of disease, and several subsequent papers have focused specifically on affective impacts including increased risk for depression, suicidal ideation, and suicide.6,7 In this issue of Cutis, Tracy et al8 provide an update on pediatric AD with an emphasis on comorbidities, quality of life, and evolving practices and therapies.
Better science, better drugs, better advocacy, better outcomes—it has not been a straight line, but it has indisputably been a forward-marching one. It is a new day, indeed.
- Siegfried EC, Jaworski JC, Eichenfield LF, et al. Developing drugs for the treatment of atopic dermatitis in children (≥3 months to <18 years of age): draft guidance for industry [published online March 30, 2018]. Pediatr Dermatol. 2018; May 35:303-322.
- Pediatric trials for AD systemic treatments. Dermatology Times. May 21, 2015. https://www.dermatologytimes.com/view/pediatric-trials-ad-systemic-treatments. Accessed August 11, 2020.
- Solimani F, Meier K, Ghoreschi K. Emerging topical and systemic JAK inhibitors in dermatology. Front Immunol. 2019;10:2847.
- Chan S, Cornelius V, Cro S, et al. Treatment effect of omalizumab on severe pediatric atopic dermatitis: the ADAPT randomized controlled trial. JAMA Pediatr. 2019;174:29-37.
- Drucker AM, Wang AR, Li W-Q, et al. The burden of atopic dermatitis: summary of a report for the National Eczema Association [published online September 8, 2016]. J Invest Dermatol. 2017;137:26-30.
- Sandhu JK, Wu KK, Bui T-L, et al. Association between atopic dermatitis and suicidality: a systematic review and meta-analysis. JAMA Dermatol. 2019;155:178-187.
- Patel KR, Immaneni S, Singam V, et al. Association between atopic dermatitis, depression, and suicidal ideation: a systematic review and meta-analysis [published online October 23, 2018]. J Am Acad Dermatol. 2019;80:402-410.
- Tracy A, Bhatti S, Eichenfield LF. Update on pediatric atopic dermatitis. Cutis. 2020;106:143-146.
- Siegfried EC, Jaworski JC, Eichenfield LF, et al. Developing drugs for the treatment of atopic dermatitis in children (≥3 months to <18 years of age): draft guidance for industry [published online March 30, 2018]. Pediatr Dermatol. 2018; May 35:303-322.
- Pediatric trials for AD systemic treatments. Dermatology Times. May 21, 2015. https://www.dermatologytimes.com/view/pediatric-trials-ad-systemic-treatments. Accessed August 11, 2020.
- Solimani F, Meier K, Ghoreschi K. Emerging topical and systemic JAK inhibitors in dermatology. Front Immunol. 2019;10:2847.
- Chan S, Cornelius V, Cro S, et al. Treatment effect of omalizumab on severe pediatric atopic dermatitis: the ADAPT randomized controlled trial. JAMA Pediatr. 2019;174:29-37.
- Drucker AM, Wang AR, Li W-Q, et al. The burden of atopic dermatitis: summary of a report for the National Eczema Association [published online September 8, 2016]. J Invest Dermatol. 2017;137:26-30.
- Sandhu JK, Wu KK, Bui T-L, et al. Association between atopic dermatitis and suicidality: a systematic review and meta-analysis. JAMA Dermatol. 2019;155:178-187.
- Patel KR, Immaneni S, Singam V, et al. Association between atopic dermatitis, depression, and suicidal ideation: a systematic review and meta-analysis [published online October 23, 2018]. J Am Acad Dermatol. 2019;80:402-410.
- Tracy A, Bhatti S, Eichenfield LF. Update on pediatric atopic dermatitis. Cutis. 2020;106:143-146.
Content Analysis of Psoriasis and Eczema Direct-to-Consumer Advertisements
Direct-to-consumer (DTC) advertisements are an important and influential source of health-related information for Americans. In 1997, the US Food and Drug Administration (FDA) relaxed regulations and permitted DTC drug advertisements to be televised. Now, via television alone, the average American is exposed to more than 30 hours annually of DTC advertisements for drugs,1 which exceeds, by far, the amount of time the average American spends with his/her physician.2 The United States spends $9.6 billion on DTC advertisements per year, of which $605 million is spent exclusively on DTC advertisements for dermatologic conditions—one of the highest amounts of spending for DTC advertisements, second only to diabetes.3
The increase in advertising for dermatologic conditions is reflective of the rapid growth in the number of treatment options available for chronic skin diseases, especially psoriasis. Since 2004, 11 biologics and 1 oral medication were FDA approved for the treatment of moderate to severe psoriasis. Despite the expansion of treatment options for psoriasis, knowledge and understanding of psoriasis and its treatments generally are poor,4,5 and undertreatment of psoriasis continues to be common.6 Data also suggest existing age and racial disparities in psoriasis treatment in the United States, whereby patients who are older or Black are less likely to receive biologic therapies.7-9 Although the exact causes of these disparities remain unclear, one study found that Black patients with psoriasis were less familiar with biologics compared to White patients,10 which suggests that the racial disparity in biologic treatment of psoriasis could be due to less exposure to and thus recognition of biologics as treatments of psoriasis among Black patients.
Some data suggest that DTC advertisements may affect drug uptake by encouraging patients to request advertised medications from their medical providers.11,12 As such, DTC advertisements are a potentially important source of exposure and information for patients. However, is it possible that DTC advertisements also may contribute to widening knowledge gaps among certain populations, and thus treatment disparities, by neglecting certain groups and targeting others with their content? In an effort to answer this question, we performed an analysis of DTC advertisements for psoriasis and eczema with special attention to advertisement placement, character representation, and disease-related content. We specifically targeted advertisements for psoriasis and eczema, as advertisements for the former are rampant and advertisements for the latter are on the rise because of emerging therapies. We hypothesized that age and racial/ethnic diversity among advertisement characters is poor, and disease-related content is lacking.
Materials and Methods
Study Design and Sample
We performed a cross-sectional analysis of televised DTC advertisements for psoriasis and eczema over 14 consecutive days (July 1, 2018, to July 14, 2018). We accessed Nielsen’s top 10 lists, specifically Prime Broadcast Network TV-United States and Prime Broadcast Programs Among African-American, from June 2018 and identified the networks with the greatest potential exposure to American consumers: ABC, CBS, FOX, and NBC.13,14 Each day, programming aired from 5
The FDA identifies DTC advertisement types as product-claim, reminder, and help-seeking advertisements. Product-claim advertisements are required to include the following information for the drug of interest: name; at least 1 FDA-approved indication; the most notable risks; and reference to a toll-free telephone number, website, or print advertisement by which a detailed summary of risks and benefits can be accessed. Reminder advertisements include the name of the drug but no information about the drug’s use.15 Help-seeking advertisements describe a disease or condition without referencing a specific drug treatment. Product-claim, reminder, and help-seeking advertisements for psoriasis or eczema that aired during the recorded time frame were included for analysis; advertisements that aired during sporting events and special programming were excluded.
DTC Advertisement Coding
Advertisement placement (ie, network, day of the week, time, associated television program), type, and target disease were documented for all advertisements included in the study. The content of each unique advertisement for psoriasis and eczema also was documented electronically in REDCap (Research Electronic Data Capture) as follows: characteristics of affected individuals and disease-related content. Advertisement coding was performed independently by 2 graduate students (A.H. and C.W.). First, one-third of the advertisements were randomly selected to be coded by both students. Intercoder agreement between the 2 students was 95.3%. Coding disagreements were primarily due to misunderstanding of definitions and were resolved through consensus. Subsequently, the remaining advertisements were randomly distributed between the 2 students, and each advertisement was coded by 1 student.
Statistical Analysis
All data were summarized descriptively with counts and frequencies using Stata 15 (StataCorp).
Results
We identified 297 DTC advertisements addressing 25 different conditions during our study period. CBS, ABC, NBC, and FOX aired 44.4%, 26.3%, 24.4%, and 5.1% of advertisements, respectively. Overall, DTC advertisements were least likely to air on Saturdays and between the hours of 5
Psoriasis DTC Advertisements
There were 5 unique psoriasis DTC advertisements, all of which were product-claim advertisements, with 1 each for secukinumab (Cosentyx [Novartis]), ixekizumab (Taltz [Eli Lilly and Company]), and guselkumab (Tremfya [Janssen Biotech, Inc]), and 2 for adalimumab (Humira [AbbVie Inc]). The advertisements aired on ABC (n=5 [38.5%]), CBS (n=5 [38.5%]), and NBC (n=3 [23.1%]). Most advertisements aired on weekdays (61.5%) between 6
Psoriasis Character Portrayal and Disease-Related Content
We identified 81 main characters who were depicted as having psoriasis among all advertisements. Characteristics of the affected characters are summarized in the Table. All affected characters were perceived to be younger adults, and there was a slight female predominance (58.0% [47/81]). Most characters were perceived to be White (92.6% [75/81]). Black and Asian characters only represented 6.2% (5/81) and 1.2% (1/81) of all affected individuals, respectively. Notably, the advertisements that featured only White main characters were aired 2.75 times more frequently than the advertisements that included non-White characters.
Psoriasis was shown on the skin of at least 1 character in an obvious depiction (ie, did not require more than 1 viewing) in 84.6% (11/13) of the advertisements. Symptoms of psoriasis (communicated either verbally or visually) were included in only 15.4% (2/13) of advertisements. No advertisements included information on the epidemiology of (ie, prevalence, subpopulations at risk), risk factors for, pathophysiology of, or comorbid diseases associated with psoriasis.
Eczema DTC Advertisements
Among the 27 eczema advertisements aired, there were 4 unique advertisements, of which 3 were product-claim advertisements (all for crisaborole [Eucrisa (Pfizer Inc)]), and 1 was a help-seeking advertisement that was sponsored by Sanofi Genzyme and Regeneron Pharmaceuticals. The advertisements aired on ABC (n=2 [7.4%]), CBS (n=17 [63.0%]), and NBC (n=8 [29.6%]). All advertisements aired on weekdays between 7
Eczema Character Portrayal and Disease-Related Content
We identified 80 main characters who were depicted to be affected by eczema among all advertisements. Characteristics of the affected characters are summarized in the Table. Most of the affected characters were perceived to be White (53.8% [43/80]) and female (71.3% [57/80]). Other races depicted included Black (28.8% [23/80]) and Asian (17.5% [14/80]). Each unique eczema advertisement included at least 1 non-White main character. Most eczema main characters were perceived to be children (66.3% [53/80]), followed by younger adults (33.8% [27/80]). No infants, teenagers, or older adults were shown as being affected by eczema.
Skin manifestations of eczema were portrayed on at least 1 character in all of the advertisements; 77.8% (21/27) of the advertisements had at least 1 obvious depiction. Symptoms of eczema and the mechanism of disease (pathophysiology) were each included in 44.4% (12/27) of advertisements. This information was included exclusively in the single help-seeking advertisement, which also referenced a website for additional disease-related information. No advertisements included information on the epidemiology of, risk factors for, or comorbid diseases associated with eczema.
Comment
In our study of televised DTC advertisements for psoriasis and eczema in the United States, we identified underrepresentation of racial/ethnic minorities and specific age groups (older adults for psoriasis and all adults for eczema) across all advertisements. Although psoriasis is suggested to be less prevalent among minority patients (1.3%–1.9% among Black patients and 1.6% among Hispanic patients) compared to White patients (2%–4%),16,17 minority vs White representation in psoriasis DTC advertisements was disproportionately lower than population-based prevalence estimates. Direct-to-consumer advertisements for eczema included more minority characters than psoriasis advertisements; however, minority representation remained inadequate considering that childhood eczema is more prevalent among Black vs White children,18 and adult eczema is at least as prevalent among minority patients compared to White patients.19 Not only was minority representation in all advertisements poor, but advertisement placement also was suboptimal, particularly for reaching Black viewers. FOX network was home to 2 of the top 3 primetime broadcast programs among Black viewers around the study period,13 yet no DTC advertisements were aired on FOX.
The current literature regarding minority representation in DTC advertisements is mixed. Some studies report underrepresentation of Black and other minority patients across a variety of diseases.20 Other studies suggest that representation of Black patients, in particular, generally is adequate, except among select serious health conditions, and that advertisements depict tokenism or stereotypical roles for minorities.21 Our study provides new and specific insight about the state of racial/ethnic and age diversity, or lack thereof, in DTC advertisements for the skin conditions that currently are most commonly targeted—psoriasis and eczema. Although it remains unclear whether DTC advertisements are good or bad, existing data suggest that potential benefits of DTC advertisements include strengthening of patient-provider relationships, reduction of underdiagnosis and undertreatment of disease, and reduction of disease stigma.22 However, in our analyses, we found disease-specific factual content among all DTC advertisements to be sparse and obvious depictions of skin disease and symptoms to be uncommon, especially for psoriasis. As such, it seems unlikely that existing DTC advertisements for psoriasis and eczema can be expected to contribute to meaningful disease education, reduce underdiagnosis, and reduce the stigmatizing attitudes that have been documented for both skin diseases.23-25
Furthermore, it is important to consider our findings in light of the role that social identity theory plays in marketing. Social identity theory supports the idea that a person’s social identity (eg, age, gender, race/ethnicity) influences his/her behavior, perceptions, and performance.26 The principle of homophily—the tendency for individuals to have positive ties to those who are similar to themselves—is a critical concept in social identity theory and suggests that consumers are more likely to pay attention to and be influenced by sources perceived as similar to themselves.20 Thus, even if the potential benefits of DTC advertisements were to be realized for psoriasis and eczema, the lack of adequate minority and older adult representation raises concerns about whether these benefits would reach a diverse population and if the advertisements might further potentiate existing knowledge and treatment disparities.
Limitations
Our study is not without limitations. The sampling period was short and might not reflect advertisement content over a longer time course. We did not evaluate other potential sources of information, such as the Internet and social media. Nevertheless, televised DTC advertisements remain a major source of medical and drug information for the general public. We did not directly evaluate viewers’ reactions to the DTC advertisements of interest; however, other literature lends support to the significance of social identity theory and its impact on consumer behavior.26
Conclusion
Our study highlights a lost opportunity among psoriasis and eczema DTC advertisements for patient reach and disease education that may encourage existing and emerging knowledge and treatment disparities for both conditions. Our findings should serve as a call to action to pharmaceutical companies and other organizations involved in creating and supporting DTC advertisements for psoriasis and eczema to increase the educational content, diversify the depicted characters, and optimize advertisement placement.
- Brownfield ED, Bernhardt JM, Phan JL, et al. Direct-to-consumer drug advertisements on network television: an exploration of quantity, frequency, and placement. J Health Commun. 2004;9:491-497.
- Tai-Seale M, McGuire TG, Zhang W. Time allocation in primary care office visits. Health Serv Res. 2007;42:1871-1894.
- Schwartz LM, Woloshin S. Medical marketing in the United States, 1997-2016. JAMA. 2019;321:80-96.
- Lanigan SW, Farber EM. Patients’ knowledge of psoriasis: pilot study. Cutis. 1990;46:359-362.
- Renzi C, Di Pietro C, Tabolli S. Participation, satisfaction and knowledge level of patients with cutaneous psoriasis or psoriatic arthritis. Clin Exp Dermatol. 2011;36:885-888.
- Lebwohl MG, Bachelez H, Barker J, et al. Patient perspectives in the management of psoriasis: results from the population-based Multinational Assessment of Psoriasis and Psoriatic Arthritis Survey. J Am Acad Dermatol. 2014;70:871-881.e871-830.
- Wu JJ, Lu M, Veverka KA, et al. The journey for US psoriasis patients prescribed a topical: a retrospective database evaluation of patient progression to oral and/or biologic treatment. J Dermatolog Treat. 2019;30:446-453.
- Takeshita J, Gelfand JM, Li P, et al. Psoriasis in the US Medicare population: prevalence, treatment, and factors associated with biologic use. J Invest Dermatol. 2015;135:2955-2963.
- Kerr GS, Qaiyumi S, Richards J, et al. Psoriasis and psoriatic arthritis in African-American patients—the need to measure disease burden. Clin Rheumatol. 2015;34:1753-1759.
- Takeshita J, Eriksen WT, Raziano VT, et al. Racial differences in perceptions of psoriasis therapies: implications for racial disparities in psoriasis treatment. J Invest Dermatol. 2019;139:1672-1679.e1.
- Wu MH, Bartz D, Avorn J, et al. Trends in direct-to-consumer advertising of prescription contraceptives. Contraception. 2016;93:398-405.
- Mintzes B, Barer ML, Kravitz RL, et al. How does direct-to-consumer advertising (DTCA) affect prescribing? a survey in primary care environments with and without legal DTCA. CMAJ. 2003;169:405-412.
- Topten. Nielson website. https://www.nielsen.com/us/en/top-ten/. Accessed July 22, 2020.
- Leading ad supported broadcast and cable networks in the United States in 2019, by average number of viewers. Statistia website. https://www.statista.com/statistics/530119/tv-networks-viewers-usa/. Accessed July 22, 2020.
- Prescription drug advertisements. Electronic Code of Federal Regulations website. https://www.ecfr.gov/cgi-bin/text-idx?SID=d4f308e364578bda8e55a831638a26c6&mc=true&node=pt21.4.202&rgn=div5. Updated August 12, 2020. Accessed August 12, 2020.
- Gelfand JM, Stern RS, Nijsten T, et al. The prevalence of psoriasis in African Americans: results from a population-based study. J Am Acad Dermatol. 2005;52:23-26.
- Rachakonda TD, Schupp CW, Armstrong AW. Psoriasis prevalence among adults in the United States. J Am Acad Dermatol. 2014;70:512-516.
- Centers for Disease Control and Prevention. National Center for Health Statistics, National Health Interview Survey, 2014. https://www.cdc.gov/nchs/data/health_policy/eczema_skin_problems_tables.pdf. Accessed July 22, 2020.
- Chiesa Fuxench ZC, Block JK, Boguniewicz M, et al. Atopic dermatitis in America study: a cross-sectional study examining the prevalence and disease burden of atopic dermatitis in the US adult population. J Invest Dermatol. 2019;139:583-590.
- Welch Cline RJ, Young HN. Marketing drugs, marketing health care relationships: a content analysis of visual cues in direct-to-consumer prescription drug advertising. Health Commun. 2004;16:131-157.
- Ball JG, Liang A, Lee WN. Representation of African Americans in direct-to-consumer pharmaceutical commercials: a content analysis with implications for health disparities. Health Mark Q. 2009;26:372-390.
- Ventola CL. Direct-to-consumer pharmaceutical advertising: therapeutic or toxic? P T. 2011;36:669-674, 681-684.
- Pearl RL, Wan MT, Takeshita J, et al. Stigmatizing attitudes toward persons with psoriasis among laypersons and medical students. J Am Acad Dermatol. 2019;80:1556-1563.
- Chernyshov PV. Stigmatization and self-perception in children with atopic dermatitis. Clin Cosmet Investig Dermatol. 2016;9:159-166.
- Wittkowski A, Richards HL, Griffiths CEM, et al. The impact of psychological and clinical factors on quality of life in individuals with atopic dermatitis. J Psychosom Res. 2004;57:195-200.
- Forehand MR, Deshpande R, Reed 2nd A. Identity salience and the influence of differential activation of the social self-schema on advertising response. J Appl Psychol. 2002;87:1086-1099.
Direct-to-consumer (DTC) advertisements are an important and influential source of health-related information for Americans. In 1997, the US Food and Drug Administration (FDA) relaxed regulations and permitted DTC drug advertisements to be televised. Now, via television alone, the average American is exposed to more than 30 hours annually of DTC advertisements for drugs,1 which exceeds, by far, the amount of time the average American spends with his/her physician.2 The United States spends $9.6 billion on DTC advertisements per year, of which $605 million is spent exclusively on DTC advertisements for dermatologic conditions—one of the highest amounts of spending for DTC advertisements, second only to diabetes.3
The increase in advertising for dermatologic conditions is reflective of the rapid growth in the number of treatment options available for chronic skin diseases, especially psoriasis. Since 2004, 11 biologics and 1 oral medication were FDA approved for the treatment of moderate to severe psoriasis. Despite the expansion of treatment options for psoriasis, knowledge and understanding of psoriasis and its treatments generally are poor,4,5 and undertreatment of psoriasis continues to be common.6 Data also suggest existing age and racial disparities in psoriasis treatment in the United States, whereby patients who are older or Black are less likely to receive biologic therapies.7-9 Although the exact causes of these disparities remain unclear, one study found that Black patients with psoriasis were less familiar with biologics compared to White patients,10 which suggests that the racial disparity in biologic treatment of psoriasis could be due to less exposure to and thus recognition of biologics as treatments of psoriasis among Black patients.
Some data suggest that DTC advertisements may affect drug uptake by encouraging patients to request advertised medications from their medical providers.11,12 As such, DTC advertisements are a potentially important source of exposure and information for patients. However, is it possible that DTC advertisements also may contribute to widening knowledge gaps among certain populations, and thus treatment disparities, by neglecting certain groups and targeting others with their content? In an effort to answer this question, we performed an analysis of DTC advertisements for psoriasis and eczema with special attention to advertisement placement, character representation, and disease-related content. We specifically targeted advertisements for psoriasis and eczema, as advertisements for the former are rampant and advertisements for the latter are on the rise because of emerging therapies. We hypothesized that age and racial/ethnic diversity among advertisement characters is poor, and disease-related content is lacking.
Materials and Methods
Study Design and Sample
We performed a cross-sectional analysis of televised DTC advertisements for psoriasis and eczema over 14 consecutive days (July 1, 2018, to July 14, 2018). We accessed Nielsen’s top 10 lists, specifically Prime Broadcast Network TV-United States and Prime Broadcast Programs Among African-American, from June 2018 and identified the networks with the greatest potential exposure to American consumers: ABC, CBS, FOX, and NBC.13,14 Each day, programming aired from 5
The FDA identifies DTC advertisement types as product-claim, reminder, and help-seeking advertisements. Product-claim advertisements are required to include the following information for the drug of interest: name; at least 1 FDA-approved indication; the most notable risks; and reference to a toll-free telephone number, website, or print advertisement by which a detailed summary of risks and benefits can be accessed. Reminder advertisements include the name of the drug but no information about the drug’s use.15 Help-seeking advertisements describe a disease or condition without referencing a specific drug treatment. Product-claim, reminder, and help-seeking advertisements for psoriasis or eczema that aired during the recorded time frame were included for analysis; advertisements that aired during sporting events and special programming were excluded.
DTC Advertisement Coding
Advertisement placement (ie, network, day of the week, time, associated television program), type, and target disease were documented for all advertisements included in the study. The content of each unique advertisement for psoriasis and eczema also was documented electronically in REDCap (Research Electronic Data Capture) as follows: characteristics of affected individuals and disease-related content. Advertisement coding was performed independently by 2 graduate students (A.H. and C.W.). First, one-third of the advertisements were randomly selected to be coded by both students. Intercoder agreement between the 2 students was 95.3%. Coding disagreements were primarily due to misunderstanding of definitions and were resolved through consensus. Subsequently, the remaining advertisements were randomly distributed between the 2 students, and each advertisement was coded by 1 student.
Statistical Analysis
All data were summarized descriptively with counts and frequencies using Stata 15 (StataCorp).
Results
We identified 297 DTC advertisements addressing 25 different conditions during our study period. CBS, ABC, NBC, and FOX aired 44.4%, 26.3%, 24.4%, and 5.1% of advertisements, respectively. Overall, DTC advertisements were least likely to air on Saturdays and between the hours of 5
Psoriasis DTC Advertisements
There were 5 unique psoriasis DTC advertisements, all of which were product-claim advertisements, with 1 each for secukinumab (Cosentyx [Novartis]), ixekizumab (Taltz [Eli Lilly and Company]), and guselkumab (Tremfya [Janssen Biotech, Inc]), and 2 for adalimumab (Humira [AbbVie Inc]). The advertisements aired on ABC (n=5 [38.5%]), CBS (n=5 [38.5%]), and NBC (n=3 [23.1%]). Most advertisements aired on weekdays (61.5%) between 6
Psoriasis Character Portrayal and Disease-Related Content
We identified 81 main characters who were depicted as having psoriasis among all advertisements. Characteristics of the affected characters are summarized in the Table. All affected characters were perceived to be younger adults, and there was a slight female predominance (58.0% [47/81]). Most characters were perceived to be White (92.6% [75/81]). Black and Asian characters only represented 6.2% (5/81) and 1.2% (1/81) of all affected individuals, respectively. Notably, the advertisements that featured only White main characters were aired 2.75 times more frequently than the advertisements that included non-White characters.
Psoriasis was shown on the skin of at least 1 character in an obvious depiction (ie, did not require more than 1 viewing) in 84.6% (11/13) of the advertisements. Symptoms of psoriasis (communicated either verbally or visually) were included in only 15.4% (2/13) of advertisements. No advertisements included information on the epidemiology of (ie, prevalence, subpopulations at risk), risk factors for, pathophysiology of, or comorbid diseases associated with psoriasis.
Eczema DTC Advertisements
Among the 27 eczema advertisements aired, there were 4 unique advertisements, of which 3 were product-claim advertisements (all for crisaborole [Eucrisa (Pfizer Inc)]), and 1 was a help-seeking advertisement that was sponsored by Sanofi Genzyme and Regeneron Pharmaceuticals. The advertisements aired on ABC (n=2 [7.4%]), CBS (n=17 [63.0%]), and NBC (n=8 [29.6%]). All advertisements aired on weekdays between 7
Eczema Character Portrayal and Disease-Related Content
We identified 80 main characters who were depicted to be affected by eczema among all advertisements. Characteristics of the affected characters are summarized in the Table. Most of the affected characters were perceived to be White (53.8% [43/80]) and female (71.3% [57/80]). Other races depicted included Black (28.8% [23/80]) and Asian (17.5% [14/80]). Each unique eczema advertisement included at least 1 non-White main character. Most eczema main characters were perceived to be children (66.3% [53/80]), followed by younger adults (33.8% [27/80]). No infants, teenagers, or older adults were shown as being affected by eczema.
Skin manifestations of eczema were portrayed on at least 1 character in all of the advertisements; 77.8% (21/27) of the advertisements had at least 1 obvious depiction. Symptoms of eczema and the mechanism of disease (pathophysiology) were each included in 44.4% (12/27) of advertisements. This information was included exclusively in the single help-seeking advertisement, which also referenced a website for additional disease-related information. No advertisements included information on the epidemiology of, risk factors for, or comorbid diseases associated with eczema.
Comment
In our study of televised DTC advertisements for psoriasis and eczema in the United States, we identified underrepresentation of racial/ethnic minorities and specific age groups (older adults for psoriasis and all adults for eczema) across all advertisements. Although psoriasis is suggested to be less prevalent among minority patients (1.3%–1.9% among Black patients and 1.6% among Hispanic patients) compared to White patients (2%–4%),16,17 minority vs White representation in psoriasis DTC advertisements was disproportionately lower than population-based prevalence estimates. Direct-to-consumer advertisements for eczema included more minority characters than psoriasis advertisements; however, minority representation remained inadequate considering that childhood eczema is more prevalent among Black vs White children,18 and adult eczema is at least as prevalent among minority patients compared to White patients.19 Not only was minority representation in all advertisements poor, but advertisement placement also was suboptimal, particularly for reaching Black viewers. FOX network was home to 2 of the top 3 primetime broadcast programs among Black viewers around the study period,13 yet no DTC advertisements were aired on FOX.
The current literature regarding minority representation in DTC advertisements is mixed. Some studies report underrepresentation of Black and other minority patients across a variety of diseases.20 Other studies suggest that representation of Black patients, in particular, generally is adequate, except among select serious health conditions, and that advertisements depict tokenism or stereotypical roles for minorities.21 Our study provides new and specific insight about the state of racial/ethnic and age diversity, or lack thereof, in DTC advertisements for the skin conditions that currently are most commonly targeted—psoriasis and eczema. Although it remains unclear whether DTC advertisements are good or bad, existing data suggest that potential benefits of DTC advertisements include strengthening of patient-provider relationships, reduction of underdiagnosis and undertreatment of disease, and reduction of disease stigma.22 However, in our analyses, we found disease-specific factual content among all DTC advertisements to be sparse and obvious depictions of skin disease and symptoms to be uncommon, especially for psoriasis. As such, it seems unlikely that existing DTC advertisements for psoriasis and eczema can be expected to contribute to meaningful disease education, reduce underdiagnosis, and reduce the stigmatizing attitudes that have been documented for both skin diseases.23-25
Furthermore, it is important to consider our findings in light of the role that social identity theory plays in marketing. Social identity theory supports the idea that a person’s social identity (eg, age, gender, race/ethnicity) influences his/her behavior, perceptions, and performance.26 The principle of homophily—the tendency for individuals to have positive ties to those who are similar to themselves—is a critical concept in social identity theory and suggests that consumers are more likely to pay attention to and be influenced by sources perceived as similar to themselves.20 Thus, even if the potential benefits of DTC advertisements were to be realized for psoriasis and eczema, the lack of adequate minority and older adult representation raises concerns about whether these benefits would reach a diverse population and if the advertisements might further potentiate existing knowledge and treatment disparities.
Limitations
Our study is not without limitations. The sampling period was short and might not reflect advertisement content over a longer time course. We did not evaluate other potential sources of information, such as the Internet and social media. Nevertheless, televised DTC advertisements remain a major source of medical and drug information for the general public. We did not directly evaluate viewers’ reactions to the DTC advertisements of interest; however, other literature lends support to the significance of social identity theory and its impact on consumer behavior.26
Conclusion
Our study highlights a lost opportunity among psoriasis and eczema DTC advertisements for patient reach and disease education that may encourage existing and emerging knowledge and treatment disparities for both conditions. Our findings should serve as a call to action to pharmaceutical companies and other organizations involved in creating and supporting DTC advertisements for psoriasis and eczema to increase the educational content, diversify the depicted characters, and optimize advertisement placement.
Direct-to-consumer (DTC) advertisements are an important and influential source of health-related information for Americans. In 1997, the US Food and Drug Administration (FDA) relaxed regulations and permitted DTC drug advertisements to be televised. Now, via television alone, the average American is exposed to more than 30 hours annually of DTC advertisements for drugs,1 which exceeds, by far, the amount of time the average American spends with his/her physician.2 The United States spends $9.6 billion on DTC advertisements per year, of which $605 million is spent exclusively on DTC advertisements for dermatologic conditions—one of the highest amounts of spending for DTC advertisements, second only to diabetes.3
The increase in advertising for dermatologic conditions is reflective of the rapid growth in the number of treatment options available for chronic skin diseases, especially psoriasis. Since 2004, 11 biologics and 1 oral medication were FDA approved for the treatment of moderate to severe psoriasis. Despite the expansion of treatment options for psoriasis, knowledge and understanding of psoriasis and its treatments generally are poor,4,5 and undertreatment of psoriasis continues to be common.6 Data also suggest existing age and racial disparities in psoriasis treatment in the United States, whereby patients who are older or Black are less likely to receive biologic therapies.7-9 Although the exact causes of these disparities remain unclear, one study found that Black patients with psoriasis were less familiar with biologics compared to White patients,10 which suggests that the racial disparity in biologic treatment of psoriasis could be due to less exposure to and thus recognition of biologics as treatments of psoriasis among Black patients.
Some data suggest that DTC advertisements may affect drug uptake by encouraging patients to request advertised medications from their medical providers.11,12 As such, DTC advertisements are a potentially important source of exposure and information for patients. However, is it possible that DTC advertisements also may contribute to widening knowledge gaps among certain populations, and thus treatment disparities, by neglecting certain groups and targeting others with their content? In an effort to answer this question, we performed an analysis of DTC advertisements for psoriasis and eczema with special attention to advertisement placement, character representation, and disease-related content. We specifically targeted advertisements for psoriasis and eczema, as advertisements for the former are rampant and advertisements for the latter are on the rise because of emerging therapies. We hypothesized that age and racial/ethnic diversity among advertisement characters is poor, and disease-related content is lacking.
Materials and Methods
Study Design and Sample
We performed a cross-sectional analysis of televised DTC advertisements for psoriasis and eczema over 14 consecutive days (July 1, 2018, to July 14, 2018). We accessed Nielsen’s top 10 lists, specifically Prime Broadcast Network TV-United States and Prime Broadcast Programs Among African-American, from June 2018 and identified the networks with the greatest potential exposure to American consumers: ABC, CBS, FOX, and NBC.13,14 Each day, programming aired from 5
The FDA identifies DTC advertisement types as product-claim, reminder, and help-seeking advertisements. Product-claim advertisements are required to include the following information for the drug of interest: name; at least 1 FDA-approved indication; the most notable risks; and reference to a toll-free telephone number, website, or print advertisement by which a detailed summary of risks and benefits can be accessed. Reminder advertisements include the name of the drug but no information about the drug’s use.15 Help-seeking advertisements describe a disease or condition without referencing a specific drug treatment. Product-claim, reminder, and help-seeking advertisements for psoriasis or eczema that aired during the recorded time frame were included for analysis; advertisements that aired during sporting events and special programming were excluded.
DTC Advertisement Coding
Advertisement placement (ie, network, day of the week, time, associated television program), type, and target disease were documented for all advertisements included in the study. The content of each unique advertisement for psoriasis and eczema also was documented electronically in REDCap (Research Electronic Data Capture) as follows: characteristics of affected individuals and disease-related content. Advertisement coding was performed independently by 2 graduate students (A.H. and C.W.). First, one-third of the advertisements were randomly selected to be coded by both students. Intercoder agreement between the 2 students was 95.3%. Coding disagreements were primarily due to misunderstanding of definitions and were resolved through consensus. Subsequently, the remaining advertisements were randomly distributed between the 2 students, and each advertisement was coded by 1 student.
Statistical Analysis
All data were summarized descriptively with counts and frequencies using Stata 15 (StataCorp).
Results
We identified 297 DTC advertisements addressing 25 different conditions during our study period. CBS, ABC, NBC, and FOX aired 44.4%, 26.3%, 24.4%, and 5.1% of advertisements, respectively. Overall, DTC advertisements were least likely to air on Saturdays and between the hours of 5
Psoriasis DTC Advertisements
There were 5 unique psoriasis DTC advertisements, all of which were product-claim advertisements, with 1 each for secukinumab (Cosentyx [Novartis]), ixekizumab (Taltz [Eli Lilly and Company]), and guselkumab (Tremfya [Janssen Biotech, Inc]), and 2 for adalimumab (Humira [AbbVie Inc]). The advertisements aired on ABC (n=5 [38.5%]), CBS (n=5 [38.5%]), and NBC (n=3 [23.1%]). Most advertisements aired on weekdays (61.5%) between 6
Psoriasis Character Portrayal and Disease-Related Content
We identified 81 main characters who were depicted as having psoriasis among all advertisements. Characteristics of the affected characters are summarized in the Table. All affected characters were perceived to be younger adults, and there was a slight female predominance (58.0% [47/81]). Most characters were perceived to be White (92.6% [75/81]). Black and Asian characters only represented 6.2% (5/81) and 1.2% (1/81) of all affected individuals, respectively. Notably, the advertisements that featured only White main characters were aired 2.75 times more frequently than the advertisements that included non-White characters.
Psoriasis was shown on the skin of at least 1 character in an obvious depiction (ie, did not require more than 1 viewing) in 84.6% (11/13) of the advertisements. Symptoms of psoriasis (communicated either verbally or visually) were included in only 15.4% (2/13) of advertisements. No advertisements included information on the epidemiology of (ie, prevalence, subpopulations at risk), risk factors for, pathophysiology of, or comorbid diseases associated with psoriasis.
Eczema DTC Advertisements
Among the 27 eczema advertisements aired, there were 4 unique advertisements, of which 3 were product-claim advertisements (all for crisaborole [Eucrisa (Pfizer Inc)]), and 1 was a help-seeking advertisement that was sponsored by Sanofi Genzyme and Regeneron Pharmaceuticals. The advertisements aired on ABC (n=2 [7.4%]), CBS (n=17 [63.0%]), and NBC (n=8 [29.6%]). All advertisements aired on weekdays between 7
Eczema Character Portrayal and Disease-Related Content
We identified 80 main characters who were depicted to be affected by eczema among all advertisements. Characteristics of the affected characters are summarized in the Table. Most of the affected characters were perceived to be White (53.8% [43/80]) and female (71.3% [57/80]). Other races depicted included Black (28.8% [23/80]) and Asian (17.5% [14/80]). Each unique eczema advertisement included at least 1 non-White main character. Most eczema main characters were perceived to be children (66.3% [53/80]), followed by younger adults (33.8% [27/80]). No infants, teenagers, or older adults were shown as being affected by eczema.
Skin manifestations of eczema were portrayed on at least 1 character in all of the advertisements; 77.8% (21/27) of the advertisements had at least 1 obvious depiction. Symptoms of eczema and the mechanism of disease (pathophysiology) were each included in 44.4% (12/27) of advertisements. This information was included exclusively in the single help-seeking advertisement, which also referenced a website for additional disease-related information. No advertisements included information on the epidemiology of, risk factors for, or comorbid diseases associated with eczema.
Comment
In our study of televised DTC advertisements for psoriasis and eczema in the United States, we identified underrepresentation of racial/ethnic minorities and specific age groups (older adults for psoriasis and all adults for eczema) across all advertisements. Although psoriasis is suggested to be less prevalent among minority patients (1.3%–1.9% among Black patients and 1.6% among Hispanic patients) compared to White patients (2%–4%),16,17 minority vs White representation in psoriasis DTC advertisements was disproportionately lower than population-based prevalence estimates. Direct-to-consumer advertisements for eczema included more minority characters than psoriasis advertisements; however, minority representation remained inadequate considering that childhood eczema is more prevalent among Black vs White children,18 and adult eczema is at least as prevalent among minority patients compared to White patients.19 Not only was minority representation in all advertisements poor, but advertisement placement also was suboptimal, particularly for reaching Black viewers. FOX network was home to 2 of the top 3 primetime broadcast programs among Black viewers around the study period,13 yet no DTC advertisements were aired on FOX.
The current literature regarding minority representation in DTC advertisements is mixed. Some studies report underrepresentation of Black and other minority patients across a variety of diseases.20 Other studies suggest that representation of Black patients, in particular, generally is adequate, except among select serious health conditions, and that advertisements depict tokenism or stereotypical roles for minorities.21 Our study provides new and specific insight about the state of racial/ethnic and age diversity, or lack thereof, in DTC advertisements for the skin conditions that currently are most commonly targeted—psoriasis and eczema. Although it remains unclear whether DTC advertisements are good or bad, existing data suggest that potential benefits of DTC advertisements include strengthening of patient-provider relationships, reduction of underdiagnosis and undertreatment of disease, and reduction of disease stigma.22 However, in our analyses, we found disease-specific factual content among all DTC advertisements to be sparse and obvious depictions of skin disease and symptoms to be uncommon, especially for psoriasis. As such, it seems unlikely that existing DTC advertisements for psoriasis and eczema can be expected to contribute to meaningful disease education, reduce underdiagnosis, and reduce the stigmatizing attitudes that have been documented for both skin diseases.23-25
Furthermore, it is important to consider our findings in light of the role that social identity theory plays in marketing. Social identity theory supports the idea that a person’s social identity (eg, age, gender, race/ethnicity) influences his/her behavior, perceptions, and performance.26 The principle of homophily—the tendency for individuals to have positive ties to those who are similar to themselves—is a critical concept in social identity theory and suggests that consumers are more likely to pay attention to and be influenced by sources perceived as similar to themselves.20 Thus, even if the potential benefits of DTC advertisements were to be realized for psoriasis and eczema, the lack of adequate minority and older adult representation raises concerns about whether these benefits would reach a diverse population and if the advertisements might further potentiate existing knowledge and treatment disparities.
Limitations
Our study is not without limitations. The sampling period was short and might not reflect advertisement content over a longer time course. We did not evaluate other potential sources of information, such as the Internet and social media. Nevertheless, televised DTC advertisements remain a major source of medical and drug information for the general public. We did not directly evaluate viewers’ reactions to the DTC advertisements of interest; however, other literature lends support to the significance of social identity theory and its impact on consumer behavior.26
Conclusion
Our study highlights a lost opportunity among psoriasis and eczema DTC advertisements for patient reach and disease education that may encourage existing and emerging knowledge and treatment disparities for both conditions. Our findings should serve as a call to action to pharmaceutical companies and other organizations involved in creating and supporting DTC advertisements for psoriasis and eczema to increase the educational content, diversify the depicted characters, and optimize advertisement placement.
- Brownfield ED, Bernhardt JM, Phan JL, et al. Direct-to-consumer drug advertisements on network television: an exploration of quantity, frequency, and placement. J Health Commun. 2004;9:491-497.
- Tai-Seale M, McGuire TG, Zhang W. Time allocation in primary care office visits. Health Serv Res. 2007;42:1871-1894.
- Schwartz LM, Woloshin S. Medical marketing in the United States, 1997-2016. JAMA. 2019;321:80-96.
- Lanigan SW, Farber EM. Patients’ knowledge of psoriasis: pilot study. Cutis. 1990;46:359-362.
- Renzi C, Di Pietro C, Tabolli S. Participation, satisfaction and knowledge level of patients with cutaneous psoriasis or psoriatic arthritis. Clin Exp Dermatol. 2011;36:885-888.
- Lebwohl MG, Bachelez H, Barker J, et al. Patient perspectives in the management of psoriasis: results from the population-based Multinational Assessment of Psoriasis and Psoriatic Arthritis Survey. J Am Acad Dermatol. 2014;70:871-881.e871-830.
- Wu JJ, Lu M, Veverka KA, et al. The journey for US psoriasis patients prescribed a topical: a retrospective database evaluation of patient progression to oral and/or biologic treatment. J Dermatolog Treat. 2019;30:446-453.
- Takeshita J, Gelfand JM, Li P, et al. Psoriasis in the US Medicare population: prevalence, treatment, and factors associated with biologic use. J Invest Dermatol. 2015;135:2955-2963.
- Kerr GS, Qaiyumi S, Richards J, et al. Psoriasis and psoriatic arthritis in African-American patients—the need to measure disease burden. Clin Rheumatol. 2015;34:1753-1759.
- Takeshita J, Eriksen WT, Raziano VT, et al. Racial differences in perceptions of psoriasis therapies: implications for racial disparities in psoriasis treatment. J Invest Dermatol. 2019;139:1672-1679.e1.
- Wu MH, Bartz D, Avorn J, et al. Trends in direct-to-consumer advertising of prescription contraceptives. Contraception. 2016;93:398-405.
- Mintzes B, Barer ML, Kravitz RL, et al. How does direct-to-consumer advertising (DTCA) affect prescribing? a survey in primary care environments with and without legal DTCA. CMAJ. 2003;169:405-412.
- Topten. Nielson website. https://www.nielsen.com/us/en/top-ten/. Accessed July 22, 2020.
- Leading ad supported broadcast and cable networks in the United States in 2019, by average number of viewers. Statistia website. https://www.statista.com/statistics/530119/tv-networks-viewers-usa/. Accessed July 22, 2020.
- Prescription drug advertisements. Electronic Code of Federal Regulations website. https://www.ecfr.gov/cgi-bin/text-idx?SID=d4f308e364578bda8e55a831638a26c6&mc=true&node=pt21.4.202&rgn=div5. Updated August 12, 2020. Accessed August 12, 2020.
- Gelfand JM, Stern RS, Nijsten T, et al. The prevalence of psoriasis in African Americans: results from a population-based study. J Am Acad Dermatol. 2005;52:23-26.
- Rachakonda TD, Schupp CW, Armstrong AW. Psoriasis prevalence among adults in the United States. J Am Acad Dermatol. 2014;70:512-516.
- Centers for Disease Control and Prevention. National Center for Health Statistics, National Health Interview Survey, 2014. https://www.cdc.gov/nchs/data/health_policy/eczema_skin_problems_tables.pdf. Accessed July 22, 2020.
- Chiesa Fuxench ZC, Block JK, Boguniewicz M, et al. Atopic dermatitis in America study: a cross-sectional study examining the prevalence and disease burden of atopic dermatitis in the US adult population. J Invest Dermatol. 2019;139:583-590.
- Welch Cline RJ, Young HN. Marketing drugs, marketing health care relationships: a content analysis of visual cues in direct-to-consumer prescription drug advertising. Health Commun. 2004;16:131-157.
- Ball JG, Liang A, Lee WN. Representation of African Americans in direct-to-consumer pharmaceutical commercials: a content analysis with implications for health disparities. Health Mark Q. 2009;26:372-390.
- Ventola CL. Direct-to-consumer pharmaceutical advertising: therapeutic or toxic? P T. 2011;36:669-674, 681-684.
- Pearl RL, Wan MT, Takeshita J, et al. Stigmatizing attitudes toward persons with psoriasis among laypersons and medical students. J Am Acad Dermatol. 2019;80:1556-1563.
- Chernyshov PV. Stigmatization and self-perception in children with atopic dermatitis. Clin Cosmet Investig Dermatol. 2016;9:159-166.
- Wittkowski A, Richards HL, Griffiths CEM, et al. The impact of psychological and clinical factors on quality of life in individuals with atopic dermatitis. J Psychosom Res. 2004;57:195-200.
- Forehand MR, Deshpande R, Reed 2nd A. Identity salience and the influence of differential activation of the social self-schema on advertising response. J Appl Psychol. 2002;87:1086-1099.
- Brownfield ED, Bernhardt JM, Phan JL, et al. Direct-to-consumer drug advertisements on network television: an exploration of quantity, frequency, and placement. J Health Commun. 2004;9:491-497.
- Tai-Seale M, McGuire TG, Zhang W. Time allocation in primary care office visits. Health Serv Res. 2007;42:1871-1894.
- Schwartz LM, Woloshin S. Medical marketing in the United States, 1997-2016. JAMA. 2019;321:80-96.
- Lanigan SW, Farber EM. Patients’ knowledge of psoriasis: pilot study. Cutis. 1990;46:359-362.
- Renzi C, Di Pietro C, Tabolli S. Participation, satisfaction and knowledge level of patients with cutaneous psoriasis or psoriatic arthritis. Clin Exp Dermatol. 2011;36:885-888.
- Lebwohl MG, Bachelez H, Barker J, et al. Patient perspectives in the management of psoriasis: results from the population-based Multinational Assessment of Psoriasis and Psoriatic Arthritis Survey. J Am Acad Dermatol. 2014;70:871-881.e871-830.
- Wu JJ, Lu M, Veverka KA, et al. The journey for US psoriasis patients prescribed a topical: a retrospective database evaluation of patient progression to oral and/or biologic treatment. J Dermatolog Treat. 2019;30:446-453.
- Takeshita J, Gelfand JM, Li P, et al. Psoriasis in the US Medicare population: prevalence, treatment, and factors associated with biologic use. J Invest Dermatol. 2015;135:2955-2963.
- Kerr GS, Qaiyumi S, Richards J, et al. Psoriasis and psoriatic arthritis in African-American patients—the need to measure disease burden. Clin Rheumatol. 2015;34:1753-1759.
- Takeshita J, Eriksen WT, Raziano VT, et al. Racial differences in perceptions of psoriasis therapies: implications for racial disparities in psoriasis treatment. J Invest Dermatol. 2019;139:1672-1679.e1.
- Wu MH, Bartz D, Avorn J, et al. Trends in direct-to-consumer advertising of prescription contraceptives. Contraception. 2016;93:398-405.
- Mintzes B, Barer ML, Kravitz RL, et al. How does direct-to-consumer advertising (DTCA) affect prescribing? a survey in primary care environments with and without legal DTCA. CMAJ. 2003;169:405-412.
- Topten. Nielson website. https://www.nielsen.com/us/en/top-ten/. Accessed July 22, 2020.
- Leading ad supported broadcast and cable networks in the United States in 2019, by average number of viewers. Statistia website. https://www.statista.com/statistics/530119/tv-networks-viewers-usa/. Accessed July 22, 2020.
- Prescription drug advertisements. Electronic Code of Federal Regulations website. https://www.ecfr.gov/cgi-bin/text-idx?SID=d4f308e364578bda8e55a831638a26c6&mc=true&node=pt21.4.202&rgn=div5. Updated August 12, 2020. Accessed August 12, 2020.
- Gelfand JM, Stern RS, Nijsten T, et al. The prevalence of psoriasis in African Americans: results from a population-based study. J Am Acad Dermatol. 2005;52:23-26.
- Rachakonda TD, Schupp CW, Armstrong AW. Psoriasis prevalence among adults in the United States. J Am Acad Dermatol. 2014;70:512-516.
- Centers for Disease Control and Prevention. National Center for Health Statistics, National Health Interview Survey, 2014. https://www.cdc.gov/nchs/data/health_policy/eczema_skin_problems_tables.pdf. Accessed July 22, 2020.
- Chiesa Fuxench ZC, Block JK, Boguniewicz M, et al. Atopic dermatitis in America study: a cross-sectional study examining the prevalence and disease burden of atopic dermatitis in the US adult population. J Invest Dermatol. 2019;139:583-590.
- Welch Cline RJ, Young HN. Marketing drugs, marketing health care relationships: a content analysis of visual cues in direct-to-consumer prescription drug advertising. Health Commun. 2004;16:131-157.
- Ball JG, Liang A, Lee WN. Representation of African Americans in direct-to-consumer pharmaceutical commercials: a content analysis with implications for health disparities. Health Mark Q. 2009;26:372-390.
- Ventola CL. Direct-to-consumer pharmaceutical advertising: therapeutic or toxic? P T. 2011;36:669-674, 681-684.
- Pearl RL, Wan MT, Takeshita J, et al. Stigmatizing attitudes toward persons with psoriasis among laypersons and medical students. J Am Acad Dermatol. 2019;80:1556-1563.
- Chernyshov PV. Stigmatization and self-perception in children with atopic dermatitis. Clin Cosmet Investig Dermatol. 2016;9:159-166.
- Wittkowski A, Richards HL, Griffiths CEM, et al. The impact of psychological and clinical factors on quality of life in individuals with atopic dermatitis. J Psychosom Res. 2004;57:195-200.
- Forehand MR, Deshpande R, Reed 2nd A. Identity salience and the influence of differential activation of the social self-schema on advertising response. J Appl Psychol. 2002;87:1086-1099.
Practice Points
- Racial/ethnic minorities and older adults are underrepresented in direct-to-consumer (DTC) advertisements for psoriasis and eczema.
- Character representation in psoriasis DTC advertisements, in particular, mirrors existing age and racial disparities in treatment with biologics.
- Disease-specific factual content was sparse, and obvious depictions of skin disease and symptoms were uncommon, especially among psoriasis DTC advertisements.
- Dermatologists should be aware of these deficiencies in psoriasis and eczema DTC advertisements and take care not to further reinforce existing knowledge gaps and inequitable treatment patterns among patients.
Update on Pediatric Atopic Dermatitis
Atopic dermatitis (AD) is a chronic, pruritic, inflammatory skin disease that occurs most frequently in children but also affects many adolescents and adults. There has been a tremendous evolution of knowledge in AD, with insights into pathogenesis, epidemiology, impact of disease, and new therapies. A variety of studies examine the epidemiology of AD and associated comorbidities. The broad developments in disease state research are reflected in new publication numbers of AD citations on PubMed. A PubMed search of articles indexed for MEDLINE at the end of 2010 using the term atopic dermatitis would have shown 965 citations during the preceding 1-year period. In the 1-year period of June 2019 to June 2020, there were more than 2000 articles. The large body of research includes work of great significance in pediatric AD, and in this article we review recent findings that are important in understanding the progress being made in the field.
Epidemiology and Comorbidities
The epidemiology of AD has evolved over the last few decades, with emerging trends and novel insights into the burden of disease.1 In a recent cross-sectional study on the epidemiology of AD in children aged 6 to 11 years, the 1-year diagnosed AD prevalence estimates worldwide included the following: United States, 10.0%; Canada, 13.3%; the EU5 Countries, 15.5%; Japan, 10.3%; and all countries studied, 12.2%.2 Another recent paper that analyzed data from the Fragile Families and Child Wellbeing Study showed that the prevalence and persistence of AD in urban US children was 15.0%.3Although pediatric AD may spontaneously remit over time, disease continuing into adolescence and adulthood is common. Paternoster et al4 studied the longitudinal course of AD in children from 2 birth cohort prospective studies, showing distinct AD phenotypes having differing course trajectories over time. Disease subsets included patients with early-onset-persistent and early-onset-late-resolving disease.4 Whether phenotyping or subgroup analysis can be used to predict disease course or risk for development of comorbidities is unknown, but it is interesting to consider how such work could influence tailoring of specific therapies to early disease presentation.
Atopic dermatitis poses a serious public health burden owing to its high prevalence, considerable morbidity and disability, increased health care utilization, and cost of care.1 Recent studies have found notably higher rates of multiple medical and mental health comorbidities in both children and adults with AD, including infections, atopic comorbidities (eg, allergic rhinitis, asthma, food allergies), eye diseases (eg, keratitis, conjunctivitis, keratoconus), and possible cardiovascular diseases and autoimmune disorders.1,5-9 Allergic comorbidities are quite common in pediatric AD patients.10 In a recent study examining the efficacy and safety of dupilumab monotherapy in 251 adolescents with moderate to severe inadequately controlled AD, most had comorbid type 2 diseases including asthma (53.6%), food allergies (60.8%), and allergic rhinitis (65.6%).11
Quality of Life/Life Impact of AD
Pediatric AD has a major impact on the quality of life of patients and their families.12 The well-being and development of children are strongly influenced by the physical and psychosocial health of parents/guardians. Two studies by Ramirez and colleagues13,14 published in 2019 examined sleep disturbances and exhaustion in mothers of children with AD. Data for the studies came from the Avon Longitudinal Study of Parents and Children. Children with active AD reported worse sleep quality than those without AD, with nearly 50% higher odds of sleep-quality disturbances. Analysis of the cohort data from 11,649 mother-child pairs who were followed up with a time-varying measure of child AD activity and severity as well as self-reported maternal sleep measures repeated at multiple time points for children aged 6 months to 11 years showed that mothers of children with AD reported difficulty falling asleep, subjectively insufficient sleep, and daytime exhaustion throughout the first 11 years of childhood.13,14 These data suggest that sleep disturbance may be a family affair.
A cross-sectional, real-world study on the burden of AD in children aged 6 to 11 years assessed by self-report demonstrated a substantial and multidimensional impact of AD, including itch, sleep disturbance, skin pain, and health-related quality-of-life impact, as well as comorbidities and school productivity losses. The burden associated with AD was remarkable and increased with disease severity.15
Drucker et al16 completed a comprehensive literature review on the burden of AD, summarized as a report for the National Eczema Association. Quality-of-life impact on pediatric patients included high rates of emotional distress; social isolation; depression; limitations in activities due to lesions with fear of triggers; and behavioral problems such as irritability, crying, and sleep disturbance resulting in difficulty performing at school.16 The psychological impact on children as well as emotional and behavioral difficulties may impact the ability for parents/guardians to implement treatment plans.17
There is a striking association between mental health disorders and AD in the US pediatric population, with a clear dose-dependent relationship that has been observed between the prevalence of a mental health disorder and the reported severity of the skin disease. Data suggest children with AD may be at increased risk for developing mental health disorders. The National Survey of Children’s Health found statistically significant increases in the likelihood of attention deficit hyperactivity disorder (odds ratio [OR], 1.87), depression (OR, 1.81), anxiety (OR, 1.77), conduct disorder (OR, 1.87), and autism (OR, 3.04).6
Evolving Practices and Therapies
Bathing Practices
There has long been much controversy regarding best bathing habits for patients with AD. In a 2009 study, cutaneous hydration was quantified after various bathing and moisturizing regimens.18 The study showed clear benefits of emollient application on skin hydration, either after bathing or without bathing. Bathing followed by emollient applications did not decrease skin hydration in contrast to bathing without emollient application.18
There are limited studies evaluating bathing frequency in pediatric patients, and many families receive conflicting information regarding best practice. In one study that surveyed 354 parents, more than 75% of parents/guardians who had seen multiple providers for their child’s AD reported a substantial amount of confusion and frustration from conflicting advice on bathing frequency.19 Cardona et al20 undertook a randomized clinical trial of frequent bathing and moisturizing vs less-frequent bathing and moisturizing in pediatric patients with AD aged 6 months to 11 years. Patients were divided into 2 groups: 1 being bathed twice daily with immediate moisturizer application and the other being bathed twice weekly followed by moisturization, then a switch to the other method. Patients used standardized topical corticosteroids (TCSs) in both groups. There were significant improvements in scoring AD and other objective measures during the frequent bathing time period vs infrequent bathing; in the group that bathed more frequently, SCORAD (SCORing Atopic Dermatitis) decreased by 21.2 compared with the group that bathed less frequently (95% confidence interval, 14.9-27.6; P<.0001). These findings suggest that more-frequent bathing with immediate moisturization is superior as an acute treatment intervention for improving AD disease severity in comparison to less-frequent bathing with immediate moisturization.20
Expanding Treatment Options
Topical Phosphodiesterase Inhibitors
There are several new and evolving topical therapies in AD. Crisaborole ointment 2% is a steroid-free phosphodiesterase inhibitor approved in 2016 by the US Food and Drug Administration (FDA) for mild to moderate AD in patients aged 2 years and older. A recent multicenter, open-label, single-arm study in 137 infants (CrisADe CARE 1) evaluated the pharmacokinetics and efficacy of crisaborole ointment 2% applied twice daily for 4 weeks in pediatric patients aged 3 months to less than 24 months of age with mild to moderate AD.21 The study had 2 cohorts: one with a minimum of 5% body surface area involvement and another (the pharmacokinetic cohort) with a minimum of 35% body surface area involvement. Both cohorts demonstrated similar efficacy data. From baseline to day 29, the mean percentage change in eczema area and severity index (EASI) score was −57.5%, and an investigator global assessment (IGA) score of clear or almost clear with at least a 2-grade improvement was achieved in 30.2% of patients. Crisaborole systemic exposures in infants were comparable with those in patients aged 2 years or older. Patients tolerated crisaborole well, with a 4% rate of burning, which was similar to other studies in children and adults but perhaps lower than seen in clinical practice. Pharmacokinetic studies did not show any remarkable noticeable concern with accumulation of propylene glycol absorption.21
Based on the CrisADe CARE 1 study data, in March 2020 the FDA extended the indication of crisaborole ointment 2% from a prior lower age limit of 24 months to approval for use in treating mild to moderate AD in children as young as 3 months, making it the first nonsteroidal topical anti-inflammatory medication to be approved in children younger than 2 years in the United States.
Evolving Topical Therapies
Topical Janus Kinase Inhibitors
Ruxolitinib is a potent inhibitor of Janus kinase 1 (JAK-1) and Janus kinase 2 (JAK-2) and has been developed in topical formulations. In recent phase 3 clinical trials of patients with AD aged 12 years and older with mild to moderate disease (TRuE-AD1 and TRuE-AD2), more than half of the patients treated with either ruxolitinib cream in a 0.75% or 1.5% concentration reached EASI-75 after 8 weeks of treatment.22 Additionally, more patients treated with topical ruxolitinib reached an IGA score of clear to almost clear than patients treated with vehicle at the end of treatment. Thus far, it appears to be very well tolerated, significantly decreases EASI score (P<.0001), and improves overall pruritus.22
Delgocitinib is a topical pan-JAK inhibitor that blocks several cytokine-signaling cascade pathways. It was first developed and approved in Japan in an ointment formulation for use in patients with AD aged 16 years and older.23 The efficacy and safety profile of delgocitinib is currently being evaluated in pediatric patients with AD in Japan. In a recent phase 2 clinical study of 103 Japanese patients aged 2 to 15 years with moderate to severe AD, patients were randomized to receive either delgocitinib ointment in 0.25% or 0.5% concentrations or vehicle ointment twice daily for 4 weeks. The proportion of patients with a modified EASI-75 score was 38.2% (13/34) in the 0.25% group and 50.0% (17/34) in the 0.5% group vs 8.6% (3/35) in the placebo group. More patients treated with delgocitinib ointment received an IGA score of clear or almost clear than patients treated with vehicle at the end of treatment. Overall, both delgocitinib groups demonstrated superior improvement in clinical symptoms and signs without notable side effects.24
Tapinarof
Tapinarof is a topical therapeutic aryl hydrocarbon receptor agonist. In a recent phase 2 randomized study of 2 concentrations and 2 frequencies of tapinarof cream vs vehicle in 247 randomized patients aged 12 to 65 years with moderate to severe disease, tapinarof demonstrated greater success with both concentrations than vehicle at all visits beyond week 2.25 Additionally, in patients treated with tapinarof cream 1%, nearly 50% reached an IGA score of clear to almost clear with at least a 2-grade improvement. More than 50% of patients achieved EASI-75 improvement at 12 weeks of treatment with tapinarof cream 1% used daily. These findings suggest that tapinarof may be an efficacious and well-tolerated treatment for both adolescents and adults with AD; however, large confirmation trials are needed to further investigate.25
Systemic Treatments
Oral JAK Inhibitors
Some of the most exciting novel therapies include several oral JAK inhibitors that target different combinations of kinases and have been shown to decrease AD severity and symptoms. Some of these agents have indications in other disease states, such as baricitinib and upadacitinib, which are both FDA approved for the treatment of rheumatoid arthritis, whereas others, such as abrocitinib, have been studied specifically for AD.
Although some agents have only been studied in adults to date, others have included adolescents in their core studies, such as abrocitinib, which received Breakthrough Therapy designation from the FDA for the treatment of patients with moderate to severe AD in February 2018. In recent phase 3 trials of patients aged 12 years and older with moderate to severe AD (JADE MONO-1 and JADE MONO-2), both doses of abrocitinib improved the IGA and EASI-75 outcomes compared with placebo.26 Additional studies will be conducted to further investigate the relative efficacy and safety in patients younger than 18 years.
Biologics
Dupilumab is a fully human monoclonal antibody that inhibits IL-4 and IL-13 signaling without suppressing the immune system. It is approved for use in patients aged 12 years and older with moderate to severe asthma and in adults with chronic rhinosinusitis with nasal polyposis. It is the first biologic to show positive results in the moderate to severe pediatric AD population. There are now extended data available exhibiting sustained benefit in adolescent patients who were continued on dupilumab therapy, evidenced by further improvement in EASI scores at the 1-year mark.27
Recently, dupilumab received approval for use in patients aged 6 to 11 years, making it the first biologic for AD to be approved for use in patients younger than 12 years. The expedited FDA approval was based on the phase 3 results in which the efficacy and safety of dupilumab combined with TCSs were compared to TCSs alone (N=367).28 In this trial, more than twice as many children achieved clear or almost clear skin and more than 4 times as many achieved itch reduction with dupilumab plus TCSs than with TCSs alone. Three-quarters of patients receiving dupilumab at the subsequently approved dosing achieved at least a 75% improvement in overall disease.28 An additional study is being conducted that includes pediatric patients aged 6 months to younger than 6 years (ClinicalTrials.gov Identifier NCT03346434).
Future Directions in Pediatric AD
Our review summarizes only some of the agents under clinical investigation for use in pediatric AD. Early treatment to establish excellent long-term disease control with aggressive topical regimens or with systemic agents may alter the course of AD and influence the development of comorbidities, though this has not yet been shown in clinical studies. The long-term impact of early treatment, along with many other intriguing issues, will be studied more in the near future.
- Silverberg JI. Public health burden and epidemiology of atopic dermatitis. Dermatol Clin. 2017;35:283-289.
- Silverberg JI, Barbarot S, Gadkari A, et al. Epidemiology of atopic dermatitis in children aged 6–11 years: a cross-sectional study in the United States (US), Canada, Europe, and Japan. Paper presented at: American Academy of Dermatology Annual Meeting; March 20-24, 2020; Denver, CO.
- McKenzie C, Silverberg JI. The prevalence and persistence of atopic dermatitis in urban United States children. Ann Allergy Asthma Immunol. 2019;123:173-178.e1.
- Paternoster L, Savenije OEM, Heron J, et al. I J Allergy Clin Immunol. 2018;141:964-971.
- Silverberg JI, Simpson EL. Association between severe eczema in children and multiple comorbid conditions and increased healthcare utilization. Pediatr Allergy Immunol. 2013;24:476-486.
- Yaghmaie P, Koudelka CW, Simpson Mental health comorbidity in patients with atopic dermatitis. J Allergy Clin Immunol. 2013;131:428-433.
- Narla S, Silverberg JI. Association between childhood atopic dermatitis and cutaneous, extracutaneous and systemic infections. Br J Dermatol. 2018;178:1467-1468.
- 2017;77:280-286. al. Incidence, prevalence, and risk of selected ocular disease in adults with atopic dermatitis. J Am Acad Dermatol.
- J Invest Dermatol. 2017;137:1074-1081. Association of atopic dermatitis with cardiovascular risk factors and diseases.
- Am J Clin Dermatol. 2018;19:821-838. Major comorbidities of atopic dermatitis: beyond allergic disorders.
- Efficacy and safety of dupilumab in adolescents with uncontrolled moderate to severe atopic dermatitis.
- Quality of life in families with children with atopic dermatitis. Pediatr Dermatol. 2016;33:28-32.
- Assessment of sleep disturbances and exhaustion in mothers of children with atopic dermatitis. JAMA Dermatol. 2019;155:556-563.
- Association of atopic dermatitis with sleep quality in children.
- Weidinger S, Simpson EL, Eckert L, et al. The patient-reported disease burden in pediatric patients with atopic dermatitis: a cross-sectional study in the United States (US), Canada, Europe, and Japan. Paperpresented at: American Academy of Dermatology Annual Meeting; March 20-24, 2020; Denver, CO.
- The burden of atopic dermatitis: summary of a report for the National Eczema Association. J Invest Dermatol. 2017;137:26-30.
- Mitchell AE. Bidirectional relationships between psychological health and dermatological conditions in children. Psychol Res Behav Manag. 2018;11:289-298.
- Chiang C, Eichenfield LF. Quantitative assessment of combination bathing and moisturizing regimens on skin hydration in atopic dermatitis. Pediatr Dermatol. 2009;26:273-278.
- Kempe E, Jain N, Cardona I. Bathing frequency recommendations for pediatric atopic dermatitis: are we adding to parental frustration? Ann Allergy Asthma Immunol. 2013;111:298‐299.
- Cardona ID, Kempe EE, Lary C, et al. Frequent versus infrequent bathing in pediatric atopic dermatitis: a randomized clinical trial. J Allergy Clin Immunol Pract. 2020;8:1014‐1021.
- , Safety, effectiveness, and pharmacokinetics of crisaborole in infants aged 3 to <24 months with mild‐to‐moderate atopic dermatitis: a phase IV open‐label study (CrisADe CARE 1). Am J Clin Dermatol. 2020;21:275-284. Gower
- Papp K, Szepietowski JC, Kircik L, et al. Efficacy and safety of ruxolitinib cream for the treatment atopic dermatitis: results from two phase 3, randomized, double-blind studies. Presented at: 2nd Annual Revolutionizing Atopic Dermatitis Conference; April 5, 2020; Chicago, IL.
- Dhillon S. Delgocitinib: first approval. Drugs. 2020;80:609‐615.
- Nakagawa H, Nemoto O, Igarashi A, et al. Phase 2 clinical study of delgocitinib ointment in pediatric patients with atopic dermatitis. J Allergy Clin Immunol. 2019;144:1575‐1583.
- Peppers J, Paller AS, Maeda-Chubachi T, et al. A phase 2, randomized dose-finding study of tapinarof (GSK2894512 cream) for the treatment of atopic dermatitis. J Am Acad Dermatol. 2019;80:89‐98.e3.
- Simpson EL, Sinclair R, Forman S, et al. Efficacy and safety of abrocitinib in adults and adolescents with moderate-to-severe atopic dermatitis (JADE MONO-1): a multicentre, double-blind, randomised, placebo-controlled, phase 3 trial. Lancet. 2020;396:255-266.
- Cork MJ, Thaçi D, Eichenfield LF, et al. Dupilumab in adolescents with uncontrolled moderate-to-severe atopic dermatitis: results from a phase IIa open-label trial and subsequent phase III open-label extension. Br J Dermatol. 2020;182:85‐96.
- Paller AS, Siegfried EC, Thaçi D, et al. Efficacy and safety of dupilumab with concomitant topical corticosteroids in children 6 to 11 years old with severe atopic dermatitis: a randomized, double-blinded, placebo-controlled phase 3 trial [published online June 20, 2020]. J Am Acad Dermatol. doi:10.1016/j.jaad.2020.06.054.
Atopic dermatitis (AD) is a chronic, pruritic, inflammatory skin disease that occurs most frequently in children but also affects many adolescents and adults. There has been a tremendous evolution of knowledge in AD, with insights into pathogenesis, epidemiology, impact of disease, and new therapies. A variety of studies examine the epidemiology of AD and associated comorbidities. The broad developments in disease state research are reflected in new publication numbers of AD citations on PubMed. A PubMed search of articles indexed for MEDLINE at the end of 2010 using the term atopic dermatitis would have shown 965 citations during the preceding 1-year period. In the 1-year period of June 2019 to June 2020, there were more than 2000 articles. The large body of research includes work of great significance in pediatric AD, and in this article we review recent findings that are important in understanding the progress being made in the field.
Epidemiology and Comorbidities
The epidemiology of AD has evolved over the last few decades, with emerging trends and novel insights into the burden of disease.1 In a recent cross-sectional study on the epidemiology of AD in children aged 6 to 11 years, the 1-year diagnosed AD prevalence estimates worldwide included the following: United States, 10.0%; Canada, 13.3%; the EU5 Countries, 15.5%; Japan, 10.3%; and all countries studied, 12.2%.2 Another recent paper that analyzed data from the Fragile Families and Child Wellbeing Study showed that the prevalence and persistence of AD in urban US children was 15.0%.3Although pediatric AD may spontaneously remit over time, disease continuing into adolescence and adulthood is common. Paternoster et al4 studied the longitudinal course of AD in children from 2 birth cohort prospective studies, showing distinct AD phenotypes having differing course trajectories over time. Disease subsets included patients with early-onset-persistent and early-onset-late-resolving disease.4 Whether phenotyping or subgroup analysis can be used to predict disease course or risk for development of comorbidities is unknown, but it is interesting to consider how such work could influence tailoring of specific therapies to early disease presentation.
Atopic dermatitis poses a serious public health burden owing to its high prevalence, considerable morbidity and disability, increased health care utilization, and cost of care.1 Recent studies have found notably higher rates of multiple medical and mental health comorbidities in both children and adults with AD, including infections, atopic comorbidities (eg, allergic rhinitis, asthma, food allergies), eye diseases (eg, keratitis, conjunctivitis, keratoconus), and possible cardiovascular diseases and autoimmune disorders.1,5-9 Allergic comorbidities are quite common in pediatric AD patients.10 In a recent study examining the efficacy and safety of dupilumab monotherapy in 251 adolescents with moderate to severe inadequately controlled AD, most had comorbid type 2 diseases including asthma (53.6%), food allergies (60.8%), and allergic rhinitis (65.6%).11
Quality of Life/Life Impact of AD
Pediatric AD has a major impact on the quality of life of patients and their families.12 The well-being and development of children are strongly influenced by the physical and psychosocial health of parents/guardians. Two studies by Ramirez and colleagues13,14 published in 2019 examined sleep disturbances and exhaustion in mothers of children with AD. Data for the studies came from the Avon Longitudinal Study of Parents and Children. Children with active AD reported worse sleep quality than those without AD, with nearly 50% higher odds of sleep-quality disturbances. Analysis of the cohort data from 11,649 mother-child pairs who were followed up with a time-varying measure of child AD activity and severity as well as self-reported maternal sleep measures repeated at multiple time points for children aged 6 months to 11 years showed that mothers of children with AD reported difficulty falling asleep, subjectively insufficient sleep, and daytime exhaustion throughout the first 11 years of childhood.13,14 These data suggest that sleep disturbance may be a family affair.
A cross-sectional, real-world study on the burden of AD in children aged 6 to 11 years assessed by self-report demonstrated a substantial and multidimensional impact of AD, including itch, sleep disturbance, skin pain, and health-related quality-of-life impact, as well as comorbidities and school productivity losses. The burden associated with AD was remarkable and increased with disease severity.15
Drucker et al16 completed a comprehensive literature review on the burden of AD, summarized as a report for the National Eczema Association. Quality-of-life impact on pediatric patients included high rates of emotional distress; social isolation; depression; limitations in activities due to lesions with fear of triggers; and behavioral problems such as irritability, crying, and sleep disturbance resulting in difficulty performing at school.16 The psychological impact on children as well as emotional and behavioral difficulties may impact the ability for parents/guardians to implement treatment plans.17
There is a striking association between mental health disorders and AD in the US pediatric population, with a clear dose-dependent relationship that has been observed between the prevalence of a mental health disorder and the reported severity of the skin disease. Data suggest children with AD may be at increased risk for developing mental health disorders. The National Survey of Children’s Health found statistically significant increases in the likelihood of attention deficit hyperactivity disorder (odds ratio [OR], 1.87), depression (OR, 1.81), anxiety (OR, 1.77), conduct disorder (OR, 1.87), and autism (OR, 3.04).6
Evolving Practices and Therapies
Bathing Practices
There has long been much controversy regarding best bathing habits for patients with AD. In a 2009 study, cutaneous hydration was quantified after various bathing and moisturizing regimens.18 The study showed clear benefits of emollient application on skin hydration, either after bathing or without bathing. Bathing followed by emollient applications did not decrease skin hydration in contrast to bathing without emollient application.18
There are limited studies evaluating bathing frequency in pediatric patients, and many families receive conflicting information regarding best practice. In one study that surveyed 354 parents, more than 75% of parents/guardians who had seen multiple providers for their child’s AD reported a substantial amount of confusion and frustration from conflicting advice on bathing frequency.19 Cardona et al20 undertook a randomized clinical trial of frequent bathing and moisturizing vs less-frequent bathing and moisturizing in pediatric patients with AD aged 6 months to 11 years. Patients were divided into 2 groups: 1 being bathed twice daily with immediate moisturizer application and the other being bathed twice weekly followed by moisturization, then a switch to the other method. Patients used standardized topical corticosteroids (TCSs) in both groups. There were significant improvements in scoring AD and other objective measures during the frequent bathing time period vs infrequent bathing; in the group that bathed more frequently, SCORAD (SCORing Atopic Dermatitis) decreased by 21.2 compared with the group that bathed less frequently (95% confidence interval, 14.9-27.6; P<.0001). These findings suggest that more-frequent bathing with immediate moisturization is superior as an acute treatment intervention for improving AD disease severity in comparison to less-frequent bathing with immediate moisturization.20
Expanding Treatment Options
Topical Phosphodiesterase Inhibitors
There are several new and evolving topical therapies in AD. Crisaborole ointment 2% is a steroid-free phosphodiesterase inhibitor approved in 2016 by the US Food and Drug Administration (FDA) for mild to moderate AD in patients aged 2 years and older. A recent multicenter, open-label, single-arm study in 137 infants (CrisADe CARE 1) evaluated the pharmacokinetics and efficacy of crisaborole ointment 2% applied twice daily for 4 weeks in pediatric patients aged 3 months to less than 24 months of age with mild to moderate AD.21 The study had 2 cohorts: one with a minimum of 5% body surface area involvement and another (the pharmacokinetic cohort) with a minimum of 35% body surface area involvement. Both cohorts demonstrated similar efficacy data. From baseline to day 29, the mean percentage change in eczema area and severity index (EASI) score was −57.5%, and an investigator global assessment (IGA) score of clear or almost clear with at least a 2-grade improvement was achieved in 30.2% of patients. Crisaborole systemic exposures in infants were comparable with those in patients aged 2 years or older. Patients tolerated crisaborole well, with a 4% rate of burning, which was similar to other studies in children and adults but perhaps lower than seen in clinical practice. Pharmacokinetic studies did not show any remarkable noticeable concern with accumulation of propylene glycol absorption.21
Based on the CrisADe CARE 1 study data, in March 2020 the FDA extended the indication of crisaborole ointment 2% from a prior lower age limit of 24 months to approval for use in treating mild to moderate AD in children as young as 3 months, making it the first nonsteroidal topical anti-inflammatory medication to be approved in children younger than 2 years in the United States.
Evolving Topical Therapies
Topical Janus Kinase Inhibitors
Ruxolitinib is a potent inhibitor of Janus kinase 1 (JAK-1) and Janus kinase 2 (JAK-2) and has been developed in topical formulations. In recent phase 3 clinical trials of patients with AD aged 12 years and older with mild to moderate disease (TRuE-AD1 and TRuE-AD2), more than half of the patients treated with either ruxolitinib cream in a 0.75% or 1.5% concentration reached EASI-75 after 8 weeks of treatment.22 Additionally, more patients treated with topical ruxolitinib reached an IGA score of clear to almost clear than patients treated with vehicle at the end of treatment. Thus far, it appears to be very well tolerated, significantly decreases EASI score (P<.0001), and improves overall pruritus.22
Delgocitinib is a topical pan-JAK inhibitor that blocks several cytokine-signaling cascade pathways. It was first developed and approved in Japan in an ointment formulation for use in patients with AD aged 16 years and older.23 The efficacy and safety profile of delgocitinib is currently being evaluated in pediatric patients with AD in Japan. In a recent phase 2 clinical study of 103 Japanese patients aged 2 to 15 years with moderate to severe AD, patients were randomized to receive either delgocitinib ointment in 0.25% or 0.5% concentrations or vehicle ointment twice daily for 4 weeks. The proportion of patients with a modified EASI-75 score was 38.2% (13/34) in the 0.25% group and 50.0% (17/34) in the 0.5% group vs 8.6% (3/35) in the placebo group. More patients treated with delgocitinib ointment received an IGA score of clear or almost clear than patients treated with vehicle at the end of treatment. Overall, both delgocitinib groups demonstrated superior improvement in clinical symptoms and signs without notable side effects.24
Tapinarof
Tapinarof is a topical therapeutic aryl hydrocarbon receptor agonist. In a recent phase 2 randomized study of 2 concentrations and 2 frequencies of tapinarof cream vs vehicle in 247 randomized patients aged 12 to 65 years with moderate to severe disease, tapinarof demonstrated greater success with both concentrations than vehicle at all visits beyond week 2.25 Additionally, in patients treated with tapinarof cream 1%, nearly 50% reached an IGA score of clear to almost clear with at least a 2-grade improvement. More than 50% of patients achieved EASI-75 improvement at 12 weeks of treatment with tapinarof cream 1% used daily. These findings suggest that tapinarof may be an efficacious and well-tolerated treatment for both adolescents and adults with AD; however, large confirmation trials are needed to further investigate.25
Systemic Treatments
Oral JAK Inhibitors
Some of the most exciting novel therapies include several oral JAK inhibitors that target different combinations of kinases and have been shown to decrease AD severity and symptoms. Some of these agents have indications in other disease states, such as baricitinib and upadacitinib, which are both FDA approved for the treatment of rheumatoid arthritis, whereas others, such as abrocitinib, have been studied specifically for AD.
Although some agents have only been studied in adults to date, others have included adolescents in their core studies, such as abrocitinib, which received Breakthrough Therapy designation from the FDA for the treatment of patients with moderate to severe AD in February 2018. In recent phase 3 trials of patients aged 12 years and older with moderate to severe AD (JADE MONO-1 and JADE MONO-2), both doses of abrocitinib improved the IGA and EASI-75 outcomes compared with placebo.26 Additional studies will be conducted to further investigate the relative efficacy and safety in patients younger than 18 years.
Biologics
Dupilumab is a fully human monoclonal antibody that inhibits IL-4 and IL-13 signaling without suppressing the immune system. It is approved for use in patients aged 12 years and older with moderate to severe asthma and in adults with chronic rhinosinusitis with nasal polyposis. It is the first biologic to show positive results in the moderate to severe pediatric AD population. There are now extended data available exhibiting sustained benefit in adolescent patients who were continued on dupilumab therapy, evidenced by further improvement in EASI scores at the 1-year mark.27
Recently, dupilumab received approval for use in patients aged 6 to 11 years, making it the first biologic for AD to be approved for use in patients younger than 12 years. The expedited FDA approval was based on the phase 3 results in which the efficacy and safety of dupilumab combined with TCSs were compared to TCSs alone (N=367).28 In this trial, more than twice as many children achieved clear or almost clear skin and more than 4 times as many achieved itch reduction with dupilumab plus TCSs than with TCSs alone. Three-quarters of patients receiving dupilumab at the subsequently approved dosing achieved at least a 75% improvement in overall disease.28 An additional study is being conducted that includes pediatric patients aged 6 months to younger than 6 years (ClinicalTrials.gov Identifier NCT03346434).
Future Directions in Pediatric AD
Our review summarizes only some of the agents under clinical investigation for use in pediatric AD. Early treatment to establish excellent long-term disease control with aggressive topical regimens or with systemic agents may alter the course of AD and influence the development of comorbidities, though this has not yet been shown in clinical studies. The long-term impact of early treatment, along with many other intriguing issues, will be studied more in the near future.
Atopic dermatitis (AD) is a chronic, pruritic, inflammatory skin disease that occurs most frequently in children but also affects many adolescents and adults. There has been a tremendous evolution of knowledge in AD, with insights into pathogenesis, epidemiology, impact of disease, and new therapies. A variety of studies examine the epidemiology of AD and associated comorbidities. The broad developments in disease state research are reflected in new publication numbers of AD citations on PubMed. A PubMed search of articles indexed for MEDLINE at the end of 2010 using the term atopic dermatitis would have shown 965 citations during the preceding 1-year period. In the 1-year period of June 2019 to June 2020, there were more than 2000 articles. The large body of research includes work of great significance in pediatric AD, and in this article we review recent findings that are important in understanding the progress being made in the field.
Epidemiology and Comorbidities
The epidemiology of AD has evolved over the last few decades, with emerging trends and novel insights into the burden of disease.1 In a recent cross-sectional study on the epidemiology of AD in children aged 6 to 11 years, the 1-year diagnosed AD prevalence estimates worldwide included the following: United States, 10.0%; Canada, 13.3%; the EU5 Countries, 15.5%; Japan, 10.3%; and all countries studied, 12.2%.2 Another recent paper that analyzed data from the Fragile Families and Child Wellbeing Study showed that the prevalence and persistence of AD in urban US children was 15.0%.3Although pediatric AD may spontaneously remit over time, disease continuing into adolescence and adulthood is common. Paternoster et al4 studied the longitudinal course of AD in children from 2 birth cohort prospective studies, showing distinct AD phenotypes having differing course trajectories over time. Disease subsets included patients with early-onset-persistent and early-onset-late-resolving disease.4 Whether phenotyping or subgroup analysis can be used to predict disease course or risk for development of comorbidities is unknown, but it is interesting to consider how such work could influence tailoring of specific therapies to early disease presentation.
Atopic dermatitis poses a serious public health burden owing to its high prevalence, considerable morbidity and disability, increased health care utilization, and cost of care.1 Recent studies have found notably higher rates of multiple medical and mental health comorbidities in both children and adults with AD, including infections, atopic comorbidities (eg, allergic rhinitis, asthma, food allergies), eye diseases (eg, keratitis, conjunctivitis, keratoconus), and possible cardiovascular diseases and autoimmune disorders.1,5-9 Allergic comorbidities are quite common in pediatric AD patients.10 In a recent study examining the efficacy and safety of dupilumab monotherapy in 251 adolescents with moderate to severe inadequately controlled AD, most had comorbid type 2 diseases including asthma (53.6%), food allergies (60.8%), and allergic rhinitis (65.6%).11
Quality of Life/Life Impact of AD
Pediatric AD has a major impact on the quality of life of patients and their families.12 The well-being and development of children are strongly influenced by the physical and psychosocial health of parents/guardians. Two studies by Ramirez and colleagues13,14 published in 2019 examined sleep disturbances and exhaustion in mothers of children with AD. Data for the studies came from the Avon Longitudinal Study of Parents and Children. Children with active AD reported worse sleep quality than those without AD, with nearly 50% higher odds of sleep-quality disturbances. Analysis of the cohort data from 11,649 mother-child pairs who were followed up with a time-varying measure of child AD activity and severity as well as self-reported maternal sleep measures repeated at multiple time points for children aged 6 months to 11 years showed that mothers of children with AD reported difficulty falling asleep, subjectively insufficient sleep, and daytime exhaustion throughout the first 11 years of childhood.13,14 These data suggest that sleep disturbance may be a family affair.
A cross-sectional, real-world study on the burden of AD in children aged 6 to 11 years assessed by self-report demonstrated a substantial and multidimensional impact of AD, including itch, sleep disturbance, skin pain, and health-related quality-of-life impact, as well as comorbidities and school productivity losses. The burden associated with AD was remarkable and increased with disease severity.15
Drucker et al16 completed a comprehensive literature review on the burden of AD, summarized as a report for the National Eczema Association. Quality-of-life impact on pediatric patients included high rates of emotional distress; social isolation; depression; limitations in activities due to lesions with fear of triggers; and behavioral problems such as irritability, crying, and sleep disturbance resulting in difficulty performing at school.16 The psychological impact on children as well as emotional and behavioral difficulties may impact the ability for parents/guardians to implement treatment plans.17
There is a striking association between mental health disorders and AD in the US pediatric population, with a clear dose-dependent relationship that has been observed between the prevalence of a mental health disorder and the reported severity of the skin disease. Data suggest children with AD may be at increased risk for developing mental health disorders. The National Survey of Children’s Health found statistically significant increases in the likelihood of attention deficit hyperactivity disorder (odds ratio [OR], 1.87), depression (OR, 1.81), anxiety (OR, 1.77), conduct disorder (OR, 1.87), and autism (OR, 3.04).6
Evolving Practices and Therapies
Bathing Practices
There has long been much controversy regarding best bathing habits for patients with AD. In a 2009 study, cutaneous hydration was quantified after various bathing and moisturizing regimens.18 The study showed clear benefits of emollient application on skin hydration, either after bathing or without bathing. Bathing followed by emollient applications did not decrease skin hydration in contrast to bathing without emollient application.18
There are limited studies evaluating bathing frequency in pediatric patients, and many families receive conflicting information regarding best practice. In one study that surveyed 354 parents, more than 75% of parents/guardians who had seen multiple providers for their child’s AD reported a substantial amount of confusion and frustration from conflicting advice on bathing frequency.19 Cardona et al20 undertook a randomized clinical trial of frequent bathing and moisturizing vs less-frequent bathing and moisturizing in pediatric patients with AD aged 6 months to 11 years. Patients were divided into 2 groups: 1 being bathed twice daily with immediate moisturizer application and the other being bathed twice weekly followed by moisturization, then a switch to the other method. Patients used standardized topical corticosteroids (TCSs) in both groups. There were significant improvements in scoring AD and other objective measures during the frequent bathing time period vs infrequent bathing; in the group that bathed more frequently, SCORAD (SCORing Atopic Dermatitis) decreased by 21.2 compared with the group that bathed less frequently (95% confidence interval, 14.9-27.6; P<.0001). These findings suggest that more-frequent bathing with immediate moisturization is superior as an acute treatment intervention for improving AD disease severity in comparison to less-frequent bathing with immediate moisturization.20
Expanding Treatment Options
Topical Phosphodiesterase Inhibitors
There are several new and evolving topical therapies in AD. Crisaborole ointment 2% is a steroid-free phosphodiesterase inhibitor approved in 2016 by the US Food and Drug Administration (FDA) for mild to moderate AD in patients aged 2 years and older. A recent multicenter, open-label, single-arm study in 137 infants (CrisADe CARE 1) evaluated the pharmacokinetics and efficacy of crisaborole ointment 2% applied twice daily for 4 weeks in pediatric patients aged 3 months to less than 24 months of age with mild to moderate AD.21 The study had 2 cohorts: one with a minimum of 5% body surface area involvement and another (the pharmacokinetic cohort) with a minimum of 35% body surface area involvement. Both cohorts demonstrated similar efficacy data. From baseline to day 29, the mean percentage change in eczema area and severity index (EASI) score was −57.5%, and an investigator global assessment (IGA) score of clear or almost clear with at least a 2-grade improvement was achieved in 30.2% of patients. Crisaborole systemic exposures in infants were comparable with those in patients aged 2 years or older. Patients tolerated crisaborole well, with a 4% rate of burning, which was similar to other studies in children and adults but perhaps lower than seen in clinical practice. Pharmacokinetic studies did not show any remarkable noticeable concern with accumulation of propylene glycol absorption.21
Based on the CrisADe CARE 1 study data, in March 2020 the FDA extended the indication of crisaborole ointment 2% from a prior lower age limit of 24 months to approval for use in treating mild to moderate AD in children as young as 3 months, making it the first nonsteroidal topical anti-inflammatory medication to be approved in children younger than 2 years in the United States.
Evolving Topical Therapies
Topical Janus Kinase Inhibitors
Ruxolitinib is a potent inhibitor of Janus kinase 1 (JAK-1) and Janus kinase 2 (JAK-2) and has been developed in topical formulations. In recent phase 3 clinical trials of patients with AD aged 12 years and older with mild to moderate disease (TRuE-AD1 and TRuE-AD2), more than half of the patients treated with either ruxolitinib cream in a 0.75% or 1.5% concentration reached EASI-75 after 8 weeks of treatment.22 Additionally, more patients treated with topical ruxolitinib reached an IGA score of clear to almost clear than patients treated with vehicle at the end of treatment. Thus far, it appears to be very well tolerated, significantly decreases EASI score (P<.0001), and improves overall pruritus.22
Delgocitinib is a topical pan-JAK inhibitor that blocks several cytokine-signaling cascade pathways. It was first developed and approved in Japan in an ointment formulation for use in patients with AD aged 16 years and older.23 The efficacy and safety profile of delgocitinib is currently being evaluated in pediatric patients with AD in Japan. In a recent phase 2 clinical study of 103 Japanese patients aged 2 to 15 years with moderate to severe AD, patients were randomized to receive either delgocitinib ointment in 0.25% or 0.5% concentrations or vehicle ointment twice daily for 4 weeks. The proportion of patients with a modified EASI-75 score was 38.2% (13/34) in the 0.25% group and 50.0% (17/34) in the 0.5% group vs 8.6% (3/35) in the placebo group. More patients treated with delgocitinib ointment received an IGA score of clear or almost clear than patients treated with vehicle at the end of treatment. Overall, both delgocitinib groups demonstrated superior improvement in clinical symptoms and signs without notable side effects.24
Tapinarof
Tapinarof is a topical therapeutic aryl hydrocarbon receptor agonist. In a recent phase 2 randomized study of 2 concentrations and 2 frequencies of tapinarof cream vs vehicle in 247 randomized patients aged 12 to 65 years with moderate to severe disease, tapinarof demonstrated greater success with both concentrations than vehicle at all visits beyond week 2.25 Additionally, in patients treated with tapinarof cream 1%, nearly 50% reached an IGA score of clear to almost clear with at least a 2-grade improvement. More than 50% of patients achieved EASI-75 improvement at 12 weeks of treatment with tapinarof cream 1% used daily. These findings suggest that tapinarof may be an efficacious and well-tolerated treatment for both adolescents and adults with AD; however, large confirmation trials are needed to further investigate.25
Systemic Treatments
Oral JAK Inhibitors
Some of the most exciting novel therapies include several oral JAK inhibitors that target different combinations of kinases and have been shown to decrease AD severity and symptoms. Some of these agents have indications in other disease states, such as baricitinib and upadacitinib, which are both FDA approved for the treatment of rheumatoid arthritis, whereas others, such as abrocitinib, have been studied specifically for AD.
Although some agents have only been studied in adults to date, others have included adolescents in their core studies, such as abrocitinib, which received Breakthrough Therapy designation from the FDA for the treatment of patients with moderate to severe AD in February 2018. In recent phase 3 trials of patients aged 12 years and older with moderate to severe AD (JADE MONO-1 and JADE MONO-2), both doses of abrocitinib improved the IGA and EASI-75 outcomes compared with placebo.26 Additional studies will be conducted to further investigate the relative efficacy and safety in patients younger than 18 years.
Biologics
Dupilumab is a fully human monoclonal antibody that inhibits IL-4 and IL-13 signaling without suppressing the immune system. It is approved for use in patients aged 12 years and older with moderate to severe asthma and in adults with chronic rhinosinusitis with nasal polyposis. It is the first biologic to show positive results in the moderate to severe pediatric AD population. There are now extended data available exhibiting sustained benefit in adolescent patients who were continued on dupilumab therapy, evidenced by further improvement in EASI scores at the 1-year mark.27
Recently, dupilumab received approval for use in patients aged 6 to 11 years, making it the first biologic for AD to be approved for use in patients younger than 12 years. The expedited FDA approval was based on the phase 3 results in which the efficacy and safety of dupilumab combined with TCSs were compared to TCSs alone (N=367).28 In this trial, more than twice as many children achieved clear or almost clear skin and more than 4 times as many achieved itch reduction with dupilumab plus TCSs than with TCSs alone. Three-quarters of patients receiving dupilumab at the subsequently approved dosing achieved at least a 75% improvement in overall disease.28 An additional study is being conducted that includes pediatric patients aged 6 months to younger than 6 years (ClinicalTrials.gov Identifier NCT03346434).
Future Directions in Pediatric AD
Our review summarizes only some of the agents under clinical investigation for use in pediatric AD. Early treatment to establish excellent long-term disease control with aggressive topical regimens or with systemic agents may alter the course of AD and influence the development of comorbidities, though this has not yet been shown in clinical studies. The long-term impact of early treatment, along with many other intriguing issues, will be studied more in the near future.
- Silverberg JI. Public health burden and epidemiology of atopic dermatitis. Dermatol Clin. 2017;35:283-289.
- Silverberg JI, Barbarot S, Gadkari A, et al. Epidemiology of atopic dermatitis in children aged 6–11 years: a cross-sectional study in the United States (US), Canada, Europe, and Japan. Paper presented at: American Academy of Dermatology Annual Meeting; March 20-24, 2020; Denver, CO.
- McKenzie C, Silverberg JI. The prevalence and persistence of atopic dermatitis in urban United States children. Ann Allergy Asthma Immunol. 2019;123:173-178.e1.
- Paternoster L, Savenije OEM, Heron J, et al. I J Allergy Clin Immunol. 2018;141:964-971.
- Silverberg JI, Simpson EL. Association between severe eczema in children and multiple comorbid conditions and increased healthcare utilization. Pediatr Allergy Immunol. 2013;24:476-486.
- Yaghmaie P, Koudelka CW, Simpson Mental health comorbidity in patients with atopic dermatitis. J Allergy Clin Immunol. 2013;131:428-433.
- Narla S, Silverberg JI. Association between childhood atopic dermatitis and cutaneous, extracutaneous and systemic infections. Br J Dermatol. 2018;178:1467-1468.
- 2017;77:280-286. al. Incidence, prevalence, and risk of selected ocular disease in adults with atopic dermatitis. J Am Acad Dermatol.
- J Invest Dermatol. 2017;137:1074-1081. Association of atopic dermatitis with cardiovascular risk factors and diseases.
- Am J Clin Dermatol. 2018;19:821-838. Major comorbidities of atopic dermatitis: beyond allergic disorders.
- Efficacy and safety of dupilumab in adolescents with uncontrolled moderate to severe atopic dermatitis.
- Quality of life in families with children with atopic dermatitis. Pediatr Dermatol. 2016;33:28-32.
- Assessment of sleep disturbances and exhaustion in mothers of children with atopic dermatitis. JAMA Dermatol. 2019;155:556-563.
- Association of atopic dermatitis with sleep quality in children.
- Weidinger S, Simpson EL, Eckert L, et al. The patient-reported disease burden in pediatric patients with atopic dermatitis: a cross-sectional study in the United States (US), Canada, Europe, and Japan. Paperpresented at: American Academy of Dermatology Annual Meeting; March 20-24, 2020; Denver, CO.
- The burden of atopic dermatitis: summary of a report for the National Eczema Association. J Invest Dermatol. 2017;137:26-30.
- Mitchell AE. Bidirectional relationships between psychological health and dermatological conditions in children. Psychol Res Behav Manag. 2018;11:289-298.
- Chiang C, Eichenfield LF. Quantitative assessment of combination bathing and moisturizing regimens on skin hydration in atopic dermatitis. Pediatr Dermatol. 2009;26:273-278.
- Kempe E, Jain N, Cardona I. Bathing frequency recommendations for pediatric atopic dermatitis: are we adding to parental frustration? Ann Allergy Asthma Immunol. 2013;111:298‐299.
- Cardona ID, Kempe EE, Lary C, et al. Frequent versus infrequent bathing in pediatric atopic dermatitis: a randomized clinical trial. J Allergy Clin Immunol Pract. 2020;8:1014‐1021.
- , Safety, effectiveness, and pharmacokinetics of crisaborole in infants aged 3 to <24 months with mild‐to‐moderate atopic dermatitis: a phase IV open‐label study (CrisADe CARE 1). Am J Clin Dermatol. 2020;21:275-284. Gower
- Papp K, Szepietowski JC, Kircik L, et al. Efficacy and safety of ruxolitinib cream for the treatment atopic dermatitis: results from two phase 3, randomized, double-blind studies. Presented at: 2nd Annual Revolutionizing Atopic Dermatitis Conference; April 5, 2020; Chicago, IL.
- Dhillon S. Delgocitinib: first approval. Drugs. 2020;80:609‐615.
- Nakagawa H, Nemoto O, Igarashi A, et al. Phase 2 clinical study of delgocitinib ointment in pediatric patients with atopic dermatitis. J Allergy Clin Immunol. 2019;144:1575‐1583.
- Peppers J, Paller AS, Maeda-Chubachi T, et al. A phase 2, randomized dose-finding study of tapinarof (GSK2894512 cream) for the treatment of atopic dermatitis. J Am Acad Dermatol. 2019;80:89‐98.e3.
- Simpson EL, Sinclair R, Forman S, et al. Efficacy and safety of abrocitinib in adults and adolescents with moderate-to-severe atopic dermatitis (JADE MONO-1): a multicentre, double-blind, randomised, placebo-controlled, phase 3 trial. Lancet. 2020;396:255-266.
- Cork MJ, Thaçi D, Eichenfield LF, et al. Dupilumab in adolescents with uncontrolled moderate-to-severe atopic dermatitis: results from a phase IIa open-label trial and subsequent phase III open-label extension. Br J Dermatol. 2020;182:85‐96.
- Paller AS, Siegfried EC, Thaçi D, et al. Efficacy and safety of dupilumab with concomitant topical corticosteroids in children 6 to 11 years old with severe atopic dermatitis: a randomized, double-blinded, placebo-controlled phase 3 trial [published online June 20, 2020]. J Am Acad Dermatol. doi:10.1016/j.jaad.2020.06.054.
- Silverberg JI. Public health burden and epidemiology of atopic dermatitis. Dermatol Clin. 2017;35:283-289.
- Silverberg JI, Barbarot S, Gadkari A, et al. Epidemiology of atopic dermatitis in children aged 6–11 years: a cross-sectional study in the United States (US), Canada, Europe, and Japan. Paper presented at: American Academy of Dermatology Annual Meeting; March 20-24, 2020; Denver, CO.
- McKenzie C, Silverberg JI. The prevalence and persistence of atopic dermatitis in urban United States children. Ann Allergy Asthma Immunol. 2019;123:173-178.e1.
- Paternoster L, Savenije OEM, Heron J, et al. I J Allergy Clin Immunol. 2018;141:964-971.
- Silverberg JI, Simpson EL. Association between severe eczema in children and multiple comorbid conditions and increased healthcare utilization. Pediatr Allergy Immunol. 2013;24:476-486.
- Yaghmaie P, Koudelka CW, Simpson Mental health comorbidity in patients with atopic dermatitis. J Allergy Clin Immunol. 2013;131:428-433.
- Narla S, Silverberg JI. Association between childhood atopic dermatitis and cutaneous, extracutaneous and systemic infections. Br J Dermatol. 2018;178:1467-1468.
- 2017;77:280-286. al. Incidence, prevalence, and risk of selected ocular disease in adults with atopic dermatitis. J Am Acad Dermatol.
- J Invest Dermatol. 2017;137:1074-1081. Association of atopic dermatitis with cardiovascular risk factors and diseases.
- Am J Clin Dermatol. 2018;19:821-838. Major comorbidities of atopic dermatitis: beyond allergic disorders.
- Efficacy and safety of dupilumab in adolescents with uncontrolled moderate to severe atopic dermatitis.
- Quality of life in families with children with atopic dermatitis. Pediatr Dermatol. 2016;33:28-32.
- Assessment of sleep disturbances and exhaustion in mothers of children with atopic dermatitis. JAMA Dermatol. 2019;155:556-563.
- Association of atopic dermatitis with sleep quality in children.
- Weidinger S, Simpson EL, Eckert L, et al. The patient-reported disease burden in pediatric patients with atopic dermatitis: a cross-sectional study in the United States (US), Canada, Europe, and Japan. Paperpresented at: American Academy of Dermatology Annual Meeting; March 20-24, 2020; Denver, CO.
- The burden of atopic dermatitis: summary of a report for the National Eczema Association. J Invest Dermatol. 2017;137:26-30.
- Mitchell AE. Bidirectional relationships between psychological health and dermatological conditions in children. Psychol Res Behav Manag. 2018;11:289-298.
- Chiang C, Eichenfield LF. Quantitative assessment of combination bathing and moisturizing regimens on skin hydration in atopic dermatitis. Pediatr Dermatol. 2009;26:273-278.
- Kempe E, Jain N, Cardona I. Bathing frequency recommendations for pediatric atopic dermatitis: are we adding to parental frustration? Ann Allergy Asthma Immunol. 2013;111:298‐299.
- Cardona ID, Kempe EE, Lary C, et al. Frequent versus infrequent bathing in pediatric atopic dermatitis: a randomized clinical trial. J Allergy Clin Immunol Pract. 2020;8:1014‐1021.
- , Safety, effectiveness, and pharmacokinetics of crisaborole in infants aged 3 to <24 months with mild‐to‐moderate atopic dermatitis: a phase IV open‐label study (CrisADe CARE 1). Am J Clin Dermatol. 2020;21:275-284. Gower
- Papp K, Szepietowski JC, Kircik L, et al. Efficacy and safety of ruxolitinib cream for the treatment atopic dermatitis: results from two phase 3, randomized, double-blind studies. Presented at: 2nd Annual Revolutionizing Atopic Dermatitis Conference; April 5, 2020; Chicago, IL.
- Dhillon S. Delgocitinib: first approval. Drugs. 2020;80:609‐615.
- Nakagawa H, Nemoto O, Igarashi A, et al. Phase 2 clinical study of delgocitinib ointment in pediatric patients with atopic dermatitis. J Allergy Clin Immunol. 2019;144:1575‐1583.
- Peppers J, Paller AS, Maeda-Chubachi T, et al. A phase 2, randomized dose-finding study of tapinarof (GSK2894512 cream) for the treatment of atopic dermatitis. J Am Acad Dermatol. 2019;80:89‐98.e3.
- Simpson EL, Sinclair R, Forman S, et al. Efficacy and safety of abrocitinib in adults and adolescents with moderate-to-severe atopic dermatitis (JADE MONO-1): a multicentre, double-blind, randomised, placebo-controlled, phase 3 trial. Lancet. 2020;396:255-266.
- Cork MJ, Thaçi D, Eichenfield LF, et al. Dupilumab in adolescents with uncontrolled moderate-to-severe atopic dermatitis: results from a phase IIa open-label trial and subsequent phase III open-label extension. Br J Dermatol. 2020;182:85‐96.
- Paller AS, Siegfried EC, Thaçi D, et al. Efficacy and safety of dupilumab with concomitant topical corticosteroids in children 6 to 11 years old with severe atopic dermatitis: a randomized, double-blinded, placebo-controlled phase 3 trial [published online June 20, 2020]. J Am Acad Dermatol. doi:10.1016/j.jaad.2020.06.054.
Practice Points
- There has been tremendous growth in our understanding of atopic dermatitis, with further insight into epidemiology, the impact on quality of life of affected individuals and their families, best bathing practices, and expanding treatment options.
- There are several novel topical and systemic agents recently approved and in late-stage clinical development programs that are evolving therapeutic approaches to pediatric disease.
Cutaneous T-cell Lymphoma and Concomitant Atopic Dermatitis Responding to Dupilumab
Patients with cutaneous T-cell lymphoma (CTCL) often are diagnosed with atopic dermatitis (AD) or psoriasis before receiving their CTCL diagnosis. The effects of new biologic therapies for AD such as dupilumab, an IL-4/IL-13 antagonist, on CTCL are unknown. Dupilumab may be beneficial in CTCL given that helper T cell (TH2) cytokines are increased in advanced CTCL.1 We present a patient with definitive CTCL and concomitant AD who was safely treated with dupilumab and experienced improvement in both CTCL and AD.
Case Report
A 68-year-old man presented with increased itching from AD and a new rash on the arms, neck, chest, back, and lower extremities (Figures 1A and 2A). He had a medical history of AD and CTCL diagnosed by biopsy and peripheral blood flow cytometry (stage IVA1 [T4N0M0B2]) that was being treated with comprehensive multimodality therapy consisting of bexarotene 375 mg daily, interferon alfa-2b 3 mIU 3 times weekly, interferon gamma-1b 2 mIU 3 times weekly, total skin electron beam therapy followed by narrowband UVB twice weekly, and extracorporeal photopheresis every 4 weeks, which resulted in a partial clinical response for 6 months. A biopsy performed at the current presentation showed focal spongiosis and features of lichen simplex chronicus with no evidence suggestive of CTCL. Peripheral blood flow cytometry showed stable B1-staged disease burden (CD4/CD8, 2.6:1); CD4+/CD7−, 12% [91/µL]; CD4+/CD26−, 21% [155/µL]). Treatment with potent and superpotent topical steroids was attempted for more than 6 months and was unsuccessful in relieving the symptoms.
Given the recalcitrant nature of the patient’s rash and itching, dupilumab was added to his CTCL regimen. Prior to initiating dupilumab, the patient reported a numeric rating scale itch intensity of 7 out of 10. After 4 weeks of treatment with dupilumab, the patient reported a numeric rating scale itch intensity of 1. Over a 3-month period, the patient’s rash improved dramatically (Figures 1B and 2B), making it possible to decrease CTCL treatments—bexarotene decreased to 300 mg, interferon alfa-2b to 3 mIU twice weekly, interferon gamma-1b to 2 mIU twice weekly, extracorporeal photopheresis every 5 weeks, and narrowband UVB was discontinued completely. A comparison of the patient’s flow cytometry analysis from before treatment to 3 months after dupilumab showed an overall slight reduction in CTCL B1 blood involvement and normalization of the patient’s absolute eosinophil count and serum lactate dehydrogenase level. The patient tolerated the treatment well without any adverse events and has maintained clinical response for 6 months.
Comment
Cutaneous T-cell lymphomas represent a heterogeneous group of T-cell lymphoproliferative disorders involving the skin.2 The definitive diagnosis of CTCL is challenging, as the clinical and pathologic features often are nonspecific in early disease. Frequently, undiagnosed patients are treated empirically with immunosuppressive agents. Tumor necrosis factor inhibitors and cyclosporine are both associated with progression or worsening of undiagnosed CTCL.3,4 Dupilumab was the first US Food and Drug Administration–approved biologic for the treatment of moderate to severe AD. Cutaneous T-cell lymphoma has immunologic features, such as TH2 skewing, that overlap with AD; however, the effects of dupilumab in CTCL are not yet known.5,6 Our group has seen patients initially thought to have AD who received dupilumab without improvement and were subsequently diagnosed with CTCL, suggesting dupilumab did not affect CTCL tumor cells. Given these findings, there was concern that dupilumab might exacerbate undiagnosed CTCL. Our patient with definitive, severe, refractory CTCL noted marked improvement in both AD and underlying CTCL with the addition of dupilumab. No other treatments were added. The response was so dramatic that we were able to wean the doses and frequencies of several CTCL treatments. Our findings suggest that dupilumab may be beneficial in a certain subset of CTCL patients with a history of AD or known concomitant AD. Prospective studies are needed to fully investigate dupilumab safety and efficacy in CTCL and whether it has any primary effects on tumor burden in addition to benefit for itch and skin symptom relief.
- Guenova E, Watanabe R, Teague JE, et al. TH2 cytokines from malignant cells suppress TH1 responses and enforce a global TH2 bias in leukemic cutaneous T-cell lymphoma. Clin Cancer Res. 2013;19:3755-3763.
- Wilcox RA. Cutaneous T-cell lymphoma: 2016 update on diagnosis, risk-stratification, and management. Am J Hematol. 2016;91:151-165.
- Martinez-Escala ME, Posligua AL, Wickless H, et al. Progression of undiagnosed cutaneous lymphoma after anti-tumor necrosis factor-alpha therapy. J Am Acad Dermatol. 2018;78:1068-1076.
- Pielop JA, Jones D, Duvic M. Transient CD30+ nodal transformation of cutaneous T-cell lymphoma associated with cyclosporine treatment. Int J Dermatol. 2001;40:505-511.
- Saulite I, Hoetzenecker W, Weidinger S, et al. Sézary syndrome and atopic dermatitis: comparison of immunological aspects and targets [published online May 17, 2016]. BioMed Res Int. doi:10.1155/2016/9717530.
- Sigurdsson V, Toonstra J, Bihari IC, et al. Interleukin 4 and interferon-gamma expression of the dermal infiltrate in patients with erythroderma and mycosis fungoides. an immuno-histochemical study. J Cutan Pathol. 2000;27:429-435.
Patients with cutaneous T-cell lymphoma (CTCL) often are diagnosed with atopic dermatitis (AD) or psoriasis before receiving their CTCL diagnosis. The effects of new biologic therapies for AD such as dupilumab, an IL-4/IL-13 antagonist, on CTCL are unknown. Dupilumab may be beneficial in CTCL given that helper T cell (TH2) cytokines are increased in advanced CTCL.1 We present a patient with definitive CTCL and concomitant AD who was safely treated with dupilumab and experienced improvement in both CTCL and AD.
Case Report
A 68-year-old man presented with increased itching from AD and a new rash on the arms, neck, chest, back, and lower extremities (Figures 1A and 2A). He had a medical history of AD and CTCL diagnosed by biopsy and peripheral blood flow cytometry (stage IVA1 [T4N0M0B2]) that was being treated with comprehensive multimodality therapy consisting of bexarotene 375 mg daily, interferon alfa-2b 3 mIU 3 times weekly, interferon gamma-1b 2 mIU 3 times weekly, total skin electron beam therapy followed by narrowband UVB twice weekly, and extracorporeal photopheresis every 4 weeks, which resulted in a partial clinical response for 6 months. A biopsy performed at the current presentation showed focal spongiosis and features of lichen simplex chronicus with no evidence suggestive of CTCL. Peripheral blood flow cytometry showed stable B1-staged disease burden (CD4/CD8, 2.6:1); CD4+/CD7−, 12% [91/µL]; CD4+/CD26−, 21% [155/µL]). Treatment with potent and superpotent topical steroids was attempted for more than 6 months and was unsuccessful in relieving the symptoms.
Given the recalcitrant nature of the patient’s rash and itching, dupilumab was added to his CTCL regimen. Prior to initiating dupilumab, the patient reported a numeric rating scale itch intensity of 7 out of 10. After 4 weeks of treatment with dupilumab, the patient reported a numeric rating scale itch intensity of 1. Over a 3-month period, the patient’s rash improved dramatically (Figures 1B and 2B), making it possible to decrease CTCL treatments—bexarotene decreased to 300 mg, interferon alfa-2b to 3 mIU twice weekly, interferon gamma-1b to 2 mIU twice weekly, extracorporeal photopheresis every 5 weeks, and narrowband UVB was discontinued completely. A comparison of the patient’s flow cytometry analysis from before treatment to 3 months after dupilumab showed an overall slight reduction in CTCL B1 blood involvement and normalization of the patient’s absolute eosinophil count and serum lactate dehydrogenase level. The patient tolerated the treatment well without any adverse events and has maintained clinical response for 6 months.
Comment
Cutaneous T-cell lymphomas represent a heterogeneous group of T-cell lymphoproliferative disorders involving the skin.2 The definitive diagnosis of CTCL is challenging, as the clinical and pathologic features often are nonspecific in early disease. Frequently, undiagnosed patients are treated empirically with immunosuppressive agents. Tumor necrosis factor inhibitors and cyclosporine are both associated with progression or worsening of undiagnosed CTCL.3,4 Dupilumab was the first US Food and Drug Administration–approved biologic for the treatment of moderate to severe AD. Cutaneous T-cell lymphoma has immunologic features, such as TH2 skewing, that overlap with AD; however, the effects of dupilumab in CTCL are not yet known.5,6 Our group has seen patients initially thought to have AD who received dupilumab without improvement and were subsequently diagnosed with CTCL, suggesting dupilumab did not affect CTCL tumor cells. Given these findings, there was concern that dupilumab might exacerbate undiagnosed CTCL. Our patient with definitive, severe, refractory CTCL noted marked improvement in both AD and underlying CTCL with the addition of dupilumab. No other treatments were added. The response was so dramatic that we were able to wean the doses and frequencies of several CTCL treatments. Our findings suggest that dupilumab may be beneficial in a certain subset of CTCL patients with a history of AD or known concomitant AD. Prospective studies are needed to fully investigate dupilumab safety and efficacy in CTCL and whether it has any primary effects on tumor burden in addition to benefit for itch and skin symptom relief.
Patients with cutaneous T-cell lymphoma (CTCL) often are diagnosed with atopic dermatitis (AD) or psoriasis before receiving their CTCL diagnosis. The effects of new biologic therapies for AD such as dupilumab, an IL-4/IL-13 antagonist, on CTCL are unknown. Dupilumab may be beneficial in CTCL given that helper T cell (TH2) cytokines are increased in advanced CTCL.1 We present a patient with definitive CTCL and concomitant AD who was safely treated with dupilumab and experienced improvement in both CTCL and AD.
Case Report
A 68-year-old man presented with increased itching from AD and a new rash on the arms, neck, chest, back, and lower extremities (Figures 1A and 2A). He had a medical history of AD and CTCL diagnosed by biopsy and peripheral blood flow cytometry (stage IVA1 [T4N0M0B2]) that was being treated with comprehensive multimodality therapy consisting of bexarotene 375 mg daily, interferon alfa-2b 3 mIU 3 times weekly, interferon gamma-1b 2 mIU 3 times weekly, total skin electron beam therapy followed by narrowband UVB twice weekly, and extracorporeal photopheresis every 4 weeks, which resulted in a partial clinical response for 6 months. A biopsy performed at the current presentation showed focal spongiosis and features of lichen simplex chronicus with no evidence suggestive of CTCL. Peripheral blood flow cytometry showed stable B1-staged disease burden (CD4/CD8, 2.6:1); CD4+/CD7−, 12% [91/µL]; CD4+/CD26−, 21% [155/µL]). Treatment with potent and superpotent topical steroids was attempted for more than 6 months and was unsuccessful in relieving the symptoms.
Given the recalcitrant nature of the patient’s rash and itching, dupilumab was added to his CTCL regimen. Prior to initiating dupilumab, the patient reported a numeric rating scale itch intensity of 7 out of 10. After 4 weeks of treatment with dupilumab, the patient reported a numeric rating scale itch intensity of 1. Over a 3-month period, the patient’s rash improved dramatically (Figures 1B and 2B), making it possible to decrease CTCL treatments—bexarotene decreased to 300 mg, interferon alfa-2b to 3 mIU twice weekly, interferon gamma-1b to 2 mIU twice weekly, extracorporeal photopheresis every 5 weeks, and narrowband UVB was discontinued completely. A comparison of the patient’s flow cytometry analysis from before treatment to 3 months after dupilumab showed an overall slight reduction in CTCL B1 blood involvement and normalization of the patient’s absolute eosinophil count and serum lactate dehydrogenase level. The patient tolerated the treatment well without any adverse events and has maintained clinical response for 6 months.
Comment
Cutaneous T-cell lymphomas represent a heterogeneous group of T-cell lymphoproliferative disorders involving the skin.2 The definitive diagnosis of CTCL is challenging, as the clinical and pathologic features often are nonspecific in early disease. Frequently, undiagnosed patients are treated empirically with immunosuppressive agents. Tumor necrosis factor inhibitors and cyclosporine are both associated with progression or worsening of undiagnosed CTCL.3,4 Dupilumab was the first US Food and Drug Administration–approved biologic for the treatment of moderate to severe AD. Cutaneous T-cell lymphoma has immunologic features, such as TH2 skewing, that overlap with AD; however, the effects of dupilumab in CTCL are not yet known.5,6 Our group has seen patients initially thought to have AD who received dupilumab without improvement and were subsequently diagnosed with CTCL, suggesting dupilumab did not affect CTCL tumor cells. Given these findings, there was concern that dupilumab might exacerbate undiagnosed CTCL. Our patient with definitive, severe, refractory CTCL noted marked improvement in both AD and underlying CTCL with the addition of dupilumab. No other treatments were added. The response was so dramatic that we were able to wean the doses and frequencies of several CTCL treatments. Our findings suggest that dupilumab may be beneficial in a certain subset of CTCL patients with a history of AD or known concomitant AD. Prospective studies are needed to fully investigate dupilumab safety and efficacy in CTCL and whether it has any primary effects on tumor burden in addition to benefit for itch and skin symptom relief.
- Guenova E, Watanabe R, Teague JE, et al. TH2 cytokines from malignant cells suppress TH1 responses and enforce a global TH2 bias in leukemic cutaneous T-cell lymphoma. Clin Cancer Res. 2013;19:3755-3763.
- Wilcox RA. Cutaneous T-cell lymphoma: 2016 update on diagnosis, risk-stratification, and management. Am J Hematol. 2016;91:151-165.
- Martinez-Escala ME, Posligua AL, Wickless H, et al. Progression of undiagnosed cutaneous lymphoma after anti-tumor necrosis factor-alpha therapy. J Am Acad Dermatol. 2018;78:1068-1076.
- Pielop JA, Jones D, Duvic M. Transient CD30+ nodal transformation of cutaneous T-cell lymphoma associated with cyclosporine treatment. Int J Dermatol. 2001;40:505-511.
- Saulite I, Hoetzenecker W, Weidinger S, et al. Sézary syndrome and atopic dermatitis: comparison of immunological aspects and targets [published online May 17, 2016]. BioMed Res Int. doi:10.1155/2016/9717530.
- Sigurdsson V, Toonstra J, Bihari IC, et al. Interleukin 4 and interferon-gamma expression of the dermal infiltrate in patients with erythroderma and mycosis fungoides. an immuno-histochemical study. J Cutan Pathol. 2000;27:429-435.
- Guenova E, Watanabe R, Teague JE, et al. TH2 cytokines from malignant cells suppress TH1 responses and enforce a global TH2 bias in leukemic cutaneous T-cell lymphoma. Clin Cancer Res. 2013;19:3755-3763.
- Wilcox RA. Cutaneous T-cell lymphoma: 2016 update on diagnosis, risk-stratification, and management. Am J Hematol. 2016;91:151-165.
- Martinez-Escala ME, Posligua AL, Wickless H, et al. Progression of undiagnosed cutaneous lymphoma after anti-tumor necrosis factor-alpha therapy. J Am Acad Dermatol. 2018;78:1068-1076.
- Pielop JA, Jones D, Duvic M. Transient CD30+ nodal transformation of cutaneous T-cell lymphoma associated with cyclosporine treatment. Int J Dermatol. 2001;40:505-511.
- Saulite I, Hoetzenecker W, Weidinger S, et al. Sézary syndrome and atopic dermatitis: comparison of immunological aspects and targets [published online May 17, 2016]. BioMed Res Int. doi:10.1155/2016/9717530.
- Sigurdsson V, Toonstra J, Bihari IC, et al. Interleukin 4 and interferon-gamma expression of the dermal infiltrate in patients with erythroderma and mycosis fungoides. an immuno-histochemical study. J Cutan Pathol. 2000;27:429-435.
Practice Points
- The diagnosis of cutaneous T-cell lymphoma (CTCL), particularly early-stage disease, remains challenging and often requires a combination of serial clinical evaluations as well as laboratory diagnostic examinations.
- Dupilumab and its effect on helper T cell (TH2) skewing may play a role in the future management of CTCL.
Smoking increases risk of high plasma NfL levels in patients with MS
A new study has found that
At the same time, patients who have stopped smoking have notably lower risk that correlates to how long ago they quit.“Before, all the studies that were looking at the association between smoking and MS – especially in terms of severity – were using indications like the Expanded Disability Status Scale and the Multiple Sclerosis Severity Score,” said first author Ali Manouchehrinia, PhD, assistant professor at the Karolinska Institute, Stockholm. “Now, we have NfL as a biomarker for disease activity, and we can see the effect of smoking on that biomarker.”
The ultimate goal, he added, “is to tease out the effects of MS severity and disease activity from NfL, to make sure that changes or differences in NfL levels are truly caused by MS and nothing else.”
Dr. Manouchehrinia presented his team’s findings at the virtual annual congress of the European Committee for Treatment and Research in Multiple Sclerosis.
To determine any associations between smoking and pNfL levels, the researchers began a retrospective, population-based cohort study of 2,572 Swedish MS patients with a self-reported history of cigarette smoking. Their average age was 38.2 years, slightly more than 71% were women, and their average disease duration was 4.13 years.
Blood samples were collected at each patients’ time of diagnosis to analyze concentrations of pNfL. Three age-stratified pNfL levels were classified: above the 80th (>C80), 95th (>C95), and 99th (>C99) of 1,026 non-MS controls’ percentiles.
Of the 2,572 MS patients, 292 (11.4%) were current regular smokers and 714 (27.8%) were past regular smokers. The past smokers’ median time since quitting was 2 years. Being a current smoker was associated with much higher odds of having pNfL levels at >C99, compared with never smokers (odds ratio, 1.52; 95% confidence interval, 1.12-2.05; P = .007) and past smokers (OR, 1.42; 95% CI, 1.01-1.99; P = .043).
For past smokers who quit between 6 and 10 years ago, the risk of having pNfL levels at >C99 was considerably lower (OR, 0.53; 95% CI, 0.29-0.93; P = .032), compared with current smokers, as was the risk for past smokers who quit more than 10 years ago (OR, 0.50; 95% CI, 0.29-0.84; P = .010). The odds were also lower, though not significantly, for patients who quit 1-5 years ago (OR, 0.84; 95% CI, 0.58-1.22; P = .359).
“It looks like, after 10 years, you go back to the baseline and have the same risk as the never smokers,” Dr. Manouchehrinia said. “But the damage may have already been done. Quitting smoking is good, but it’s better to not smoke at all.”
Dr. Manouchehrinia acknowledged the study’s limitations, including the need to learn more about the role NfL levels – especially plasma NfL levels – play across MS patients, along with the complications surrounding smoking as an environmental factor. He noted that, in Sweden, many people get their nicotine from snuff rather than cigarettes. “Among our MS population, we’ve seen a recent shift toward female snuff users,” which lessens the amount they smoke and could confound the results. In fact, the study indicated that snuff users had less risk of pNfL levels at >C95, compared with nonsnuff users (OR, 0.71; 95% CI, 0.51-0.97; P = .034).
The authors reported several potential conflicts of interest, including receiving research grants and lecture honoraria and serving on advisory boards for various pharmaceutical companies.
A new study has found that
At the same time, patients who have stopped smoking have notably lower risk that correlates to how long ago they quit.“Before, all the studies that were looking at the association between smoking and MS – especially in terms of severity – were using indications like the Expanded Disability Status Scale and the Multiple Sclerosis Severity Score,” said first author Ali Manouchehrinia, PhD, assistant professor at the Karolinska Institute, Stockholm. “Now, we have NfL as a biomarker for disease activity, and we can see the effect of smoking on that biomarker.”
The ultimate goal, he added, “is to tease out the effects of MS severity and disease activity from NfL, to make sure that changes or differences in NfL levels are truly caused by MS and nothing else.”
Dr. Manouchehrinia presented his team’s findings at the virtual annual congress of the European Committee for Treatment and Research in Multiple Sclerosis.
To determine any associations between smoking and pNfL levels, the researchers began a retrospective, population-based cohort study of 2,572 Swedish MS patients with a self-reported history of cigarette smoking. Their average age was 38.2 years, slightly more than 71% were women, and their average disease duration was 4.13 years.
Blood samples were collected at each patients’ time of diagnosis to analyze concentrations of pNfL. Three age-stratified pNfL levels were classified: above the 80th (>C80), 95th (>C95), and 99th (>C99) of 1,026 non-MS controls’ percentiles.
Of the 2,572 MS patients, 292 (11.4%) were current regular smokers and 714 (27.8%) were past regular smokers. The past smokers’ median time since quitting was 2 years. Being a current smoker was associated with much higher odds of having pNfL levels at >C99, compared with never smokers (odds ratio, 1.52; 95% confidence interval, 1.12-2.05; P = .007) and past smokers (OR, 1.42; 95% CI, 1.01-1.99; P = .043).
For past smokers who quit between 6 and 10 years ago, the risk of having pNfL levels at >C99 was considerably lower (OR, 0.53; 95% CI, 0.29-0.93; P = .032), compared with current smokers, as was the risk for past smokers who quit more than 10 years ago (OR, 0.50; 95% CI, 0.29-0.84; P = .010). The odds were also lower, though not significantly, for patients who quit 1-5 years ago (OR, 0.84; 95% CI, 0.58-1.22; P = .359).
“It looks like, after 10 years, you go back to the baseline and have the same risk as the never smokers,” Dr. Manouchehrinia said. “But the damage may have already been done. Quitting smoking is good, but it’s better to not smoke at all.”
Dr. Manouchehrinia acknowledged the study’s limitations, including the need to learn more about the role NfL levels – especially plasma NfL levels – play across MS patients, along with the complications surrounding smoking as an environmental factor. He noted that, in Sweden, many people get their nicotine from snuff rather than cigarettes. “Among our MS population, we’ve seen a recent shift toward female snuff users,” which lessens the amount they smoke and could confound the results. In fact, the study indicated that snuff users had less risk of pNfL levels at >C95, compared with nonsnuff users (OR, 0.71; 95% CI, 0.51-0.97; P = .034).
The authors reported several potential conflicts of interest, including receiving research grants and lecture honoraria and serving on advisory boards for various pharmaceutical companies.
A new study has found that
At the same time, patients who have stopped smoking have notably lower risk that correlates to how long ago they quit.“Before, all the studies that were looking at the association between smoking and MS – especially in terms of severity – were using indications like the Expanded Disability Status Scale and the Multiple Sclerosis Severity Score,” said first author Ali Manouchehrinia, PhD, assistant professor at the Karolinska Institute, Stockholm. “Now, we have NfL as a biomarker for disease activity, and we can see the effect of smoking on that biomarker.”
The ultimate goal, he added, “is to tease out the effects of MS severity and disease activity from NfL, to make sure that changes or differences in NfL levels are truly caused by MS and nothing else.”
Dr. Manouchehrinia presented his team’s findings at the virtual annual congress of the European Committee for Treatment and Research in Multiple Sclerosis.
To determine any associations between smoking and pNfL levels, the researchers began a retrospective, population-based cohort study of 2,572 Swedish MS patients with a self-reported history of cigarette smoking. Their average age was 38.2 years, slightly more than 71% were women, and their average disease duration was 4.13 years.
Blood samples were collected at each patients’ time of diagnosis to analyze concentrations of pNfL. Three age-stratified pNfL levels were classified: above the 80th (>C80), 95th (>C95), and 99th (>C99) of 1,026 non-MS controls’ percentiles.
Of the 2,572 MS patients, 292 (11.4%) were current regular smokers and 714 (27.8%) were past regular smokers. The past smokers’ median time since quitting was 2 years. Being a current smoker was associated with much higher odds of having pNfL levels at >C99, compared with never smokers (odds ratio, 1.52; 95% confidence interval, 1.12-2.05; P = .007) and past smokers (OR, 1.42; 95% CI, 1.01-1.99; P = .043).
For past smokers who quit between 6 and 10 years ago, the risk of having pNfL levels at >C99 was considerably lower (OR, 0.53; 95% CI, 0.29-0.93; P = .032), compared with current smokers, as was the risk for past smokers who quit more than 10 years ago (OR, 0.50; 95% CI, 0.29-0.84; P = .010). The odds were also lower, though not significantly, for patients who quit 1-5 years ago (OR, 0.84; 95% CI, 0.58-1.22; P = .359).
“It looks like, after 10 years, you go back to the baseline and have the same risk as the never smokers,” Dr. Manouchehrinia said. “But the damage may have already been done. Quitting smoking is good, but it’s better to not smoke at all.”
Dr. Manouchehrinia acknowledged the study’s limitations, including the need to learn more about the role NfL levels – especially plasma NfL levels – play across MS patients, along with the complications surrounding smoking as an environmental factor. He noted that, in Sweden, many people get their nicotine from snuff rather than cigarettes. “Among our MS population, we’ve seen a recent shift toward female snuff users,” which lessens the amount they smoke and could confound the results. In fact, the study indicated that snuff users had less risk of pNfL levels at >C95, compared with nonsnuff users (OR, 0.71; 95% CI, 0.51-0.97; P = .034).
The authors reported several potential conflicts of interest, including receiving research grants and lecture honoraria and serving on advisory boards for various pharmaceutical companies.
FROM ECTRIMS 2020