User login
Melanoma in Hispanics: We May Have It All Wrong
To the Editor:
We read with interest the commentary by Srivastava et al,1 "The Dayanara Effect: Increasing Skin Cancer Awareness in the Hispanic Community," concerning former Miss Universe Dayanara Torres and her diagnosis of metastatic melanoma; however, we believe it misses the mark. A quick Google search shows that Ms. Torres has fair skin and blue eyes. She has lived most of her life in Puerto Rico, the Philippines, and California--places where sun exposure is high and may have contributed to her diagnosis. Factors that have been linked to an increased risk for melanoma are fair skin, red or blonde hair, blue or green eyes, intense intermittent sun exposure and sunburns, a weakened immune system, and a family history of skin cancer.2 Although we do not know her complete medical history, Ms. Torres' skin phenotype and likely chronic UV exposure made her a candidate for skin cancer. Although Srivastava et al1 acknowledged that the Hispanic community encompasses a wide variety of individuals with varying levels of skin pigmentation and sun sensitivity, they overlooked Ms. Torres' risk for skin cancer because of her ethnic background. This form of generalization may negatively affect patient care and safety. By 2060, Hispanics are projected to account for almost 30% of the US population,3 and we must acknowledge the flaws that exist in our overall methodology for assessing skin cancer risk among this population to provide patients with unbiased care.
In the early 1970s, the United States adopted the ethnonym Hispanic as a way of conglomerating Spanish-speaking individuals from Spain, the Caribbean, and Central and South America.4 The goal was to implement a common identifier that enabled the US Government to study the economic and social development of these groups. Nevertheless, considerable differences exist among distinct Hispanic communities, and variations in skin pigmentation and sun sensitivity are no exception. Although Hispanic countries are an amalgam of diverse races due to colonization, some have stronger European, African, or Amerindian influences, limiting the use of ethnicity during melanoma risk assessment. Another misconception reflected in the commentary by Srivastava et al1 is the belief that the terms white and Hispanic are mutually exclusive. A study examining melanoma rates in the Chilean population supports this claim.5 The genetic composition of the Chilean high socioeconomic strata is 5% Amerindian and 95% white, while the low socioeconomic strata is approximately 40% Amerindian and 60% white. Patients from the low socioeconomic strata had higher rates of acral malignant melanoma, which typically is seen in patients with skin of color. On the other hand, males from the high socioeconomic strata had higher rates of truncal melanoma, which is more common among the white population.5 These results suggest that while both groups are considered Hispanic, it is ancestral origin that contributes to the differential rates and types of malignant melanoma.
When analyzing data regarding melanoma rates in Hispanics, particularly data collected in the United States, we must question if the results are representative of the entire population. One point worth emphasizing is that melanoma data in the Hispanic community often is flawed. The North American Association of Central Cancer Registries considers Europeans such as Spaniards, as well as citizens of Andorra, the Canary Islands, and the Balearic Islands as Hispanic.6 Additionally, the Florida Cancer Data System uses data such as country of birth, ethnicity, and surname or maiden name recorded by the hospital tumor registry to identify Hispanic patients with melanoma.7 In 2006, Hu et al7 used the Florida Cancer Data System to analyze melanoma data in Miami-Dade County in South Florida, which has the second largest Hispanic community in the United States. One limitation to such data is that ethnicity often is self-reported by patients or assigned by a health care provider. In addition, women whose maiden names are not available may be misclassified through marriage depending on whether their husbands have Spanish or non-Spanish last names.7 Finally, with societal norms evolving, Americans are now more accepting of interracial marriages. In 2017, the Pew Research Center reported that 17% of all newlyweds in the United States were intermarried and 42% of these marriages were between a white individual and a Hispanic individual, comprising the most prevalent form of intermarriage reported.8 In 2015, 27% of newlywed Hispanics were intermarried. This percentage varied depending on whether they were born in the United States or abroad. Although 15% of Hispanic immigrants married a spouse from another race, 39% of Hispanics born in the United States married a non-Hispanic (eg, white, black, Asian, or Native American who is not Hispanic).8 This type of marriage and subsequent offspring might lead to an increase in the white genetic pool. As a result, the risk for melanoma development may be increased or misrepresented. Remaining aware of these changes in the population is crucial, as it exemplifies why the current methodology for gathering and reporting melanoma data is unreliable.
Labeling Ms. Torres as Hispanic due to her Puerto Rican nationality did not tell us anything about her risk for developing melanoma. To correctly assess the risk for melanoma among Hispanics, it is imperative that we re-evaluate our approach. We agree with He et al9 that our efforts should be dedicated to better understanding the impact of pigmentation, race, genetics, and sunburn on the risk for melanoma. Until we know more about this possible correlation, we should reconsider how we study melanoma using Hispanics as an ethnicity. We may have it all wrong.
- Srivastava R, Wassef C, Rao BK. The Dayanara effect: increasing skin cancer awareness in the Hispanic community. Cutis. 2019;103:257-258.
- Curiel-Lewandrowski C. Risk factors for the development of melanoma. UpToDate. https://www.uptodate.com/contents/risk-factors-for-the-development-of-melanoma. Updated February 27, 2020. Accessed April 16, 2020.
- Colby SL, Ortman JM. Projections of the size and composition of the U.S. population: 2014 to 2060. United States Census Bureau website. https://www.census.gov/library/publications/2015/demo/p25-1143.html. Published March 3, 2015. Accessed April 16, 2020.
- National Research Council (US) Panel on Hispanics in the United States; Tienda M, Mitchell F, editors. Multiple Origins, Uncertain Destinies: Hispanics and the American Future. Washington (DC): National Academies Press (US); 2006. 3, Defining Hispanicity: E Pluribus Unum or E Pluribus Plures? Available from: https://www.ncbi.nlm.nih.gov/books/NBK19811/
- Zemelman VB, Valenzuela CY, Sazunic I, et al. Malignant melanoma in Chile: different site distribution between private and state patients. Biol Res. 2014;47:34.
- NAACCR Race and Ethnicity Work Group. NAACCR guideline for enhancing Hispanic-Latino identification: revised NAACCR Hispanic/Latino identification algorithm [NHIA v2.2.1]. NAACCR website. https://www.naaccr.org/wp-content/uploads/2016/11/NHIA_v2_2_1_09122011.pdf. Revised September 12, 2011. Accessed April 15, 2020.
- Hu S, Soza-Vento RM, Parker DF, et al. Comparison of stage at diagnosis of melanoma among Hispanic, black, and white patients in Miami-Dade County, Florida. Arch Dermatol. 2006;142:704-708.
- Livingston G, Brown A. Intermarriage in the U.S. 50 years after Loving v. Virginia. Pew Research Center website. https://www.pewsocialtrends.org/2017/05/18/intermarriage-in-the-u-s-50-years-after-loving-v-virginia/. Published May 18, 2017. Accessed April 15, 2020.
- He SY, McCulloch CE, Boscardin WJ, et al. Self-reported pigmentary phenotypes and race are significant but incomplete predictors of Fitzpatrick skin phototype in an ethnically diverse population. J Am Acad Dermatol. 2014;71:731-737.
Authors' Response
While Ms. Cruzval-O'Reilly and Dr. Lugo-Somolinos highlight many important points on conducting meaningful research for the Hispanic community, they seem to have misunderstood the overall purpose of our commentary,1 which was to highlight the increased skin cancer awareness that a notable and vocal member of the Hispanic community brought to our academic dermatology clinic, rather than to discuss skin types within the Hispanic community. As the authors mentioned, the term Hispanic is a descriptor of ethnicity rather than race, and Hispanic patients may have varying levels of skin pigmentation and sun sensitivity. While Dayanara Torres may have risk factors for developing melanoma, minimizing her connection with the Hispanic community because of her fair skin and light eyes would be a mistake. It not only isolates members of the Hispanic community that are of skin types I and II, but it also discounts the power of her story and language in raising awareness. We observed an increase in Hispanic patients presenting to our clinic who were concerned about skin cancer after Ms. Torres shared her diagnosis of metastatic melanoma through social media, followed by Spanish language educational videos on melanoma.
Several studies have described disparities among Hispanic patients diagnosed with melanoma as compared to their non-Hispanic white counterparts, including younger age at diagnosis, later stage of presentation, increased presence of regional involvement, and worse mortality.2-6 Furthermore, a small study of high school students by Ma et al7 showed disparities in skin cancer knowledge, perceived risk, and sun-protective behaviors among Hispanic whites and non-Hispanic whites, which remained significant (P<.05) after controlling for skin pigmentation and sun sensitivity. We agree with the authors that further analysis of skin type, race, genetics, and other risk factors may help refine the research on skin cancer disparities within the Hispanic community. We suspect that disparities may persist even when examining these factors. There have been several studies showing that knowledge-based interventions, especially when delivered in Spanish, improve understanding of skin cancer, personal risk, and self-examinations, and we support Ms. Torres' efforts in utilizing her platform to provide information about melanoma in Spanish.8-12
Radhika Srivastava, MD; Cindy Wassef, MD; Babar K. Rao, MD
From the Department of Dermatology, Rutgers Robert Wood Johnson Medical School, Somerset, New Jersey.
The authors report no conflict of interest.
Correspondence: Radhika Srivastava, MD, 1 World's Fair Dr, Ste 2400, Somerset, NJ 08873 ([email protected]).
References
- Srivastava R, Wassef C, Rao BK. The Dayanara effect: increasing skin cancer awareness in the Hispanic community. Cutis. 2019;103:257-258.
- Perez MI. Skin cancer in Hispanics in the United States. J Drugs Dermatol. 2019;18:s117-s120.
- Higgins S, Nazemi A, Feinstein S, et al. Clinical presentations of melanoma in African Americans, Hispanics, and Asians. Dermatol Surg. 2019;45:791-801.
- Harvey VM, Oldfield CW, Chen JT, et al. Melanoma disparities among US Hispanics: use of the social ecological model to contextualize reasons for inequitable outcomes and frame a research agenda [published online August 29, 2016]. J Skin Cancer. doi:10.1155/2016/4635740
- Garnett E, Townsend J, Steele B, et al. Characteristics, rates, and trends of melanoma incidence among Hispanics in the USA. Cancer Causes Control. 2016;27:647-659.
- Rouhani P, Hu S, Kirsner RS. Melanoma in Hispanic and black Americans. Cancer Control. 2008;15:248-253.
- Ma F, Collado-Mesa F, Hu S, et al. Skin cancer awareness and sun protection behaviors in white Hispanic and white non-Hispanic high school students in Miami, Florida. Arch Dermatol. 2007;143:983-988.
- Kundu RV, Kamaria M, Ortiz S, et al. Effectiveness of a knowledge-based intervention for melanoma among those with ethnic skin. J Am Acad Dermatol. 2010;62:777-784.
- Kailas A, Botwin AL, Pritchett EN, et al. Assessing the effectiveness of knowledge-based interventions in increasing skin cancer awareness, knowledge, and protective behaviors in skin of color populations. Cutis. 2017;100:235-240.
- Roman CJ, Guan X, Barnholtz-Sloan J, et al. A trial online educational melanoma program aimed at the Hispanic population improves knowledge and behaviors. Dermatol Surg. 2016;42:672-676.
- Hernandez C, Kim H, Mauleon G, et al. A pilot program in collaboration with community centers to increase awareness and participation in skin cancer screening among Latinos in Chicago. J Cancer Educ. 2013;28:342-345.
- Chung GY, Brown G, Gibson D. Increasing melanoma screening among Hispanic/Latino Americans: a community-based educational intervention. Health Educ Behav. 2015;42:627-632.
To the Editor:
We read with interest the commentary by Srivastava et al,1 "The Dayanara Effect: Increasing Skin Cancer Awareness in the Hispanic Community," concerning former Miss Universe Dayanara Torres and her diagnosis of metastatic melanoma; however, we believe it misses the mark. A quick Google search shows that Ms. Torres has fair skin and blue eyes. She has lived most of her life in Puerto Rico, the Philippines, and California--places where sun exposure is high and may have contributed to her diagnosis. Factors that have been linked to an increased risk for melanoma are fair skin, red or blonde hair, blue or green eyes, intense intermittent sun exposure and sunburns, a weakened immune system, and a family history of skin cancer.2 Although we do not know her complete medical history, Ms. Torres' skin phenotype and likely chronic UV exposure made her a candidate for skin cancer. Although Srivastava et al1 acknowledged that the Hispanic community encompasses a wide variety of individuals with varying levels of skin pigmentation and sun sensitivity, they overlooked Ms. Torres' risk for skin cancer because of her ethnic background. This form of generalization may negatively affect patient care and safety. By 2060, Hispanics are projected to account for almost 30% of the US population,3 and we must acknowledge the flaws that exist in our overall methodology for assessing skin cancer risk among this population to provide patients with unbiased care.
In the early 1970s, the United States adopted the ethnonym Hispanic as a way of conglomerating Spanish-speaking individuals from Spain, the Caribbean, and Central and South America.4 The goal was to implement a common identifier that enabled the US Government to study the economic and social development of these groups. Nevertheless, considerable differences exist among distinct Hispanic communities, and variations in skin pigmentation and sun sensitivity are no exception. Although Hispanic countries are an amalgam of diverse races due to colonization, some have stronger European, African, or Amerindian influences, limiting the use of ethnicity during melanoma risk assessment. Another misconception reflected in the commentary by Srivastava et al1 is the belief that the terms white and Hispanic are mutually exclusive. A study examining melanoma rates in the Chilean population supports this claim.5 The genetic composition of the Chilean high socioeconomic strata is 5% Amerindian and 95% white, while the low socioeconomic strata is approximately 40% Amerindian and 60% white. Patients from the low socioeconomic strata had higher rates of acral malignant melanoma, which typically is seen in patients with skin of color. On the other hand, males from the high socioeconomic strata had higher rates of truncal melanoma, which is more common among the white population.5 These results suggest that while both groups are considered Hispanic, it is ancestral origin that contributes to the differential rates and types of malignant melanoma.
When analyzing data regarding melanoma rates in Hispanics, particularly data collected in the United States, we must question if the results are representative of the entire population. One point worth emphasizing is that melanoma data in the Hispanic community often is flawed. The North American Association of Central Cancer Registries considers Europeans such as Spaniards, as well as citizens of Andorra, the Canary Islands, and the Balearic Islands as Hispanic.6 Additionally, the Florida Cancer Data System uses data such as country of birth, ethnicity, and surname or maiden name recorded by the hospital tumor registry to identify Hispanic patients with melanoma.7 In 2006, Hu et al7 used the Florida Cancer Data System to analyze melanoma data in Miami-Dade County in South Florida, which has the second largest Hispanic community in the United States. One limitation to such data is that ethnicity often is self-reported by patients or assigned by a health care provider. In addition, women whose maiden names are not available may be misclassified through marriage depending on whether their husbands have Spanish or non-Spanish last names.7 Finally, with societal norms evolving, Americans are now more accepting of interracial marriages. In 2017, the Pew Research Center reported that 17% of all newlyweds in the United States were intermarried and 42% of these marriages were between a white individual and a Hispanic individual, comprising the most prevalent form of intermarriage reported.8 In 2015, 27% of newlywed Hispanics were intermarried. This percentage varied depending on whether they were born in the United States or abroad. Although 15% of Hispanic immigrants married a spouse from another race, 39% of Hispanics born in the United States married a non-Hispanic (eg, white, black, Asian, or Native American who is not Hispanic).8 This type of marriage and subsequent offspring might lead to an increase in the white genetic pool. As a result, the risk for melanoma development may be increased or misrepresented. Remaining aware of these changes in the population is crucial, as it exemplifies why the current methodology for gathering and reporting melanoma data is unreliable.
Labeling Ms. Torres as Hispanic due to her Puerto Rican nationality did not tell us anything about her risk for developing melanoma. To correctly assess the risk for melanoma among Hispanics, it is imperative that we re-evaluate our approach. We agree with He et al9 that our efforts should be dedicated to better understanding the impact of pigmentation, race, genetics, and sunburn on the risk for melanoma. Until we know more about this possible correlation, we should reconsider how we study melanoma using Hispanics as an ethnicity. We may have it all wrong.
- Srivastava R, Wassef C, Rao BK. The Dayanara effect: increasing skin cancer awareness in the Hispanic community. Cutis. 2019;103:257-258.
- Curiel-Lewandrowski C. Risk factors for the development of melanoma. UpToDate. https://www.uptodate.com/contents/risk-factors-for-the-development-of-melanoma. Updated February 27, 2020. Accessed April 16, 2020.
- Colby SL, Ortman JM. Projections of the size and composition of the U.S. population: 2014 to 2060. United States Census Bureau website. https://www.census.gov/library/publications/2015/demo/p25-1143.html. Published March 3, 2015. Accessed April 16, 2020.
- National Research Council (US) Panel on Hispanics in the United States; Tienda M, Mitchell F, editors. Multiple Origins, Uncertain Destinies: Hispanics and the American Future. Washington (DC): National Academies Press (US); 2006. 3, Defining Hispanicity: E Pluribus Unum or E Pluribus Plures? Available from: https://www.ncbi.nlm.nih.gov/books/NBK19811/
- Zemelman VB, Valenzuela CY, Sazunic I, et al. Malignant melanoma in Chile: different site distribution between private and state patients. Biol Res. 2014;47:34.
- NAACCR Race and Ethnicity Work Group. NAACCR guideline for enhancing Hispanic-Latino identification: revised NAACCR Hispanic/Latino identification algorithm [NHIA v2.2.1]. NAACCR website. https://www.naaccr.org/wp-content/uploads/2016/11/NHIA_v2_2_1_09122011.pdf. Revised September 12, 2011. Accessed April 15, 2020.
- Hu S, Soza-Vento RM, Parker DF, et al. Comparison of stage at diagnosis of melanoma among Hispanic, black, and white patients in Miami-Dade County, Florida. Arch Dermatol. 2006;142:704-708.
- Livingston G, Brown A. Intermarriage in the U.S. 50 years after Loving v. Virginia. Pew Research Center website. https://www.pewsocialtrends.org/2017/05/18/intermarriage-in-the-u-s-50-years-after-loving-v-virginia/. Published May 18, 2017. Accessed April 15, 2020.
- He SY, McCulloch CE, Boscardin WJ, et al. Self-reported pigmentary phenotypes and race are significant but incomplete predictors of Fitzpatrick skin phototype in an ethnically diverse population. J Am Acad Dermatol. 2014;71:731-737.
Authors' Response
While Ms. Cruzval-O'Reilly and Dr. Lugo-Somolinos highlight many important points on conducting meaningful research for the Hispanic community, they seem to have misunderstood the overall purpose of our commentary,1 which was to highlight the increased skin cancer awareness that a notable and vocal member of the Hispanic community brought to our academic dermatology clinic, rather than to discuss skin types within the Hispanic community. As the authors mentioned, the term Hispanic is a descriptor of ethnicity rather than race, and Hispanic patients may have varying levels of skin pigmentation and sun sensitivity. While Dayanara Torres may have risk factors for developing melanoma, minimizing her connection with the Hispanic community because of her fair skin and light eyes would be a mistake. It not only isolates members of the Hispanic community that are of skin types I and II, but it also discounts the power of her story and language in raising awareness. We observed an increase in Hispanic patients presenting to our clinic who were concerned about skin cancer after Ms. Torres shared her diagnosis of metastatic melanoma through social media, followed by Spanish language educational videos on melanoma.
Several studies have described disparities among Hispanic patients diagnosed with melanoma as compared to their non-Hispanic white counterparts, including younger age at diagnosis, later stage of presentation, increased presence of regional involvement, and worse mortality.2-6 Furthermore, a small study of high school students by Ma et al7 showed disparities in skin cancer knowledge, perceived risk, and sun-protective behaviors among Hispanic whites and non-Hispanic whites, which remained significant (P<.05) after controlling for skin pigmentation and sun sensitivity. We agree with the authors that further analysis of skin type, race, genetics, and other risk factors may help refine the research on skin cancer disparities within the Hispanic community. We suspect that disparities may persist even when examining these factors. There have been several studies showing that knowledge-based interventions, especially when delivered in Spanish, improve understanding of skin cancer, personal risk, and self-examinations, and we support Ms. Torres' efforts in utilizing her platform to provide information about melanoma in Spanish.8-12
Radhika Srivastava, MD; Cindy Wassef, MD; Babar K. Rao, MD
From the Department of Dermatology, Rutgers Robert Wood Johnson Medical School, Somerset, New Jersey.
The authors report no conflict of interest.
Correspondence: Radhika Srivastava, MD, 1 World's Fair Dr, Ste 2400, Somerset, NJ 08873 ([email protected]).
References
- Srivastava R, Wassef C, Rao BK. The Dayanara effect: increasing skin cancer awareness in the Hispanic community. Cutis. 2019;103:257-258.
- Perez MI. Skin cancer in Hispanics in the United States. J Drugs Dermatol. 2019;18:s117-s120.
- Higgins S, Nazemi A, Feinstein S, et al. Clinical presentations of melanoma in African Americans, Hispanics, and Asians. Dermatol Surg. 2019;45:791-801.
- Harvey VM, Oldfield CW, Chen JT, et al. Melanoma disparities among US Hispanics: use of the social ecological model to contextualize reasons for inequitable outcomes and frame a research agenda [published online August 29, 2016]. J Skin Cancer. doi:10.1155/2016/4635740
- Garnett E, Townsend J, Steele B, et al. Characteristics, rates, and trends of melanoma incidence among Hispanics in the USA. Cancer Causes Control. 2016;27:647-659.
- Rouhani P, Hu S, Kirsner RS. Melanoma in Hispanic and black Americans. Cancer Control. 2008;15:248-253.
- Ma F, Collado-Mesa F, Hu S, et al. Skin cancer awareness and sun protection behaviors in white Hispanic and white non-Hispanic high school students in Miami, Florida. Arch Dermatol. 2007;143:983-988.
- Kundu RV, Kamaria M, Ortiz S, et al. Effectiveness of a knowledge-based intervention for melanoma among those with ethnic skin. J Am Acad Dermatol. 2010;62:777-784.
- Kailas A, Botwin AL, Pritchett EN, et al. Assessing the effectiveness of knowledge-based interventions in increasing skin cancer awareness, knowledge, and protective behaviors in skin of color populations. Cutis. 2017;100:235-240.
- Roman CJ, Guan X, Barnholtz-Sloan J, et al. A trial online educational melanoma program aimed at the Hispanic population improves knowledge and behaviors. Dermatol Surg. 2016;42:672-676.
- Hernandez C, Kim H, Mauleon G, et al. A pilot program in collaboration with community centers to increase awareness and participation in skin cancer screening among Latinos in Chicago. J Cancer Educ. 2013;28:342-345.
- Chung GY, Brown G, Gibson D. Increasing melanoma screening among Hispanic/Latino Americans: a community-based educational intervention. Health Educ Behav. 2015;42:627-632.
To the Editor:
We read with interest the commentary by Srivastava et al,1 "The Dayanara Effect: Increasing Skin Cancer Awareness in the Hispanic Community," concerning former Miss Universe Dayanara Torres and her diagnosis of metastatic melanoma; however, we believe it misses the mark. A quick Google search shows that Ms. Torres has fair skin and blue eyes. She has lived most of her life in Puerto Rico, the Philippines, and California--places where sun exposure is high and may have contributed to her diagnosis. Factors that have been linked to an increased risk for melanoma are fair skin, red or blonde hair, blue or green eyes, intense intermittent sun exposure and sunburns, a weakened immune system, and a family history of skin cancer.2 Although we do not know her complete medical history, Ms. Torres' skin phenotype and likely chronic UV exposure made her a candidate for skin cancer. Although Srivastava et al1 acknowledged that the Hispanic community encompasses a wide variety of individuals with varying levels of skin pigmentation and sun sensitivity, they overlooked Ms. Torres' risk for skin cancer because of her ethnic background. This form of generalization may negatively affect patient care and safety. By 2060, Hispanics are projected to account for almost 30% of the US population,3 and we must acknowledge the flaws that exist in our overall methodology for assessing skin cancer risk among this population to provide patients with unbiased care.
In the early 1970s, the United States adopted the ethnonym Hispanic as a way of conglomerating Spanish-speaking individuals from Spain, the Caribbean, and Central and South America.4 The goal was to implement a common identifier that enabled the US Government to study the economic and social development of these groups. Nevertheless, considerable differences exist among distinct Hispanic communities, and variations in skin pigmentation and sun sensitivity are no exception. Although Hispanic countries are an amalgam of diverse races due to colonization, some have stronger European, African, or Amerindian influences, limiting the use of ethnicity during melanoma risk assessment. Another misconception reflected in the commentary by Srivastava et al1 is the belief that the terms white and Hispanic are mutually exclusive. A study examining melanoma rates in the Chilean population supports this claim.5 The genetic composition of the Chilean high socioeconomic strata is 5% Amerindian and 95% white, while the low socioeconomic strata is approximately 40% Amerindian and 60% white. Patients from the low socioeconomic strata had higher rates of acral malignant melanoma, which typically is seen in patients with skin of color. On the other hand, males from the high socioeconomic strata had higher rates of truncal melanoma, which is more common among the white population.5 These results suggest that while both groups are considered Hispanic, it is ancestral origin that contributes to the differential rates and types of malignant melanoma.
When analyzing data regarding melanoma rates in Hispanics, particularly data collected in the United States, we must question if the results are representative of the entire population. One point worth emphasizing is that melanoma data in the Hispanic community often is flawed. The North American Association of Central Cancer Registries considers Europeans such as Spaniards, as well as citizens of Andorra, the Canary Islands, and the Balearic Islands as Hispanic.6 Additionally, the Florida Cancer Data System uses data such as country of birth, ethnicity, and surname or maiden name recorded by the hospital tumor registry to identify Hispanic patients with melanoma.7 In 2006, Hu et al7 used the Florida Cancer Data System to analyze melanoma data in Miami-Dade County in South Florida, which has the second largest Hispanic community in the United States. One limitation to such data is that ethnicity often is self-reported by patients or assigned by a health care provider. In addition, women whose maiden names are not available may be misclassified through marriage depending on whether their husbands have Spanish or non-Spanish last names.7 Finally, with societal norms evolving, Americans are now more accepting of interracial marriages. In 2017, the Pew Research Center reported that 17% of all newlyweds in the United States were intermarried and 42% of these marriages were between a white individual and a Hispanic individual, comprising the most prevalent form of intermarriage reported.8 In 2015, 27% of newlywed Hispanics were intermarried. This percentage varied depending on whether they were born in the United States or abroad. Although 15% of Hispanic immigrants married a spouse from another race, 39% of Hispanics born in the United States married a non-Hispanic (eg, white, black, Asian, or Native American who is not Hispanic).8 This type of marriage and subsequent offspring might lead to an increase in the white genetic pool. As a result, the risk for melanoma development may be increased or misrepresented. Remaining aware of these changes in the population is crucial, as it exemplifies why the current methodology for gathering and reporting melanoma data is unreliable.
Labeling Ms. Torres as Hispanic due to her Puerto Rican nationality did not tell us anything about her risk for developing melanoma. To correctly assess the risk for melanoma among Hispanics, it is imperative that we re-evaluate our approach. We agree with He et al9 that our efforts should be dedicated to better understanding the impact of pigmentation, race, genetics, and sunburn on the risk for melanoma. Until we know more about this possible correlation, we should reconsider how we study melanoma using Hispanics as an ethnicity. We may have it all wrong.
- Srivastava R, Wassef C, Rao BK. The Dayanara effect: increasing skin cancer awareness in the Hispanic community. Cutis. 2019;103:257-258.
- Curiel-Lewandrowski C. Risk factors for the development of melanoma. UpToDate. https://www.uptodate.com/contents/risk-factors-for-the-development-of-melanoma. Updated February 27, 2020. Accessed April 16, 2020.
- Colby SL, Ortman JM. Projections of the size and composition of the U.S. population: 2014 to 2060. United States Census Bureau website. https://www.census.gov/library/publications/2015/demo/p25-1143.html. Published March 3, 2015. Accessed April 16, 2020.
- National Research Council (US) Panel on Hispanics in the United States; Tienda M, Mitchell F, editors. Multiple Origins, Uncertain Destinies: Hispanics and the American Future. Washington (DC): National Academies Press (US); 2006. 3, Defining Hispanicity: E Pluribus Unum or E Pluribus Plures? Available from: https://www.ncbi.nlm.nih.gov/books/NBK19811/
- Zemelman VB, Valenzuela CY, Sazunic I, et al. Malignant melanoma in Chile: different site distribution between private and state patients. Biol Res. 2014;47:34.
- NAACCR Race and Ethnicity Work Group. NAACCR guideline for enhancing Hispanic-Latino identification: revised NAACCR Hispanic/Latino identification algorithm [NHIA v2.2.1]. NAACCR website. https://www.naaccr.org/wp-content/uploads/2016/11/NHIA_v2_2_1_09122011.pdf. Revised September 12, 2011. Accessed April 15, 2020.
- Hu S, Soza-Vento RM, Parker DF, et al. Comparison of stage at diagnosis of melanoma among Hispanic, black, and white patients in Miami-Dade County, Florida. Arch Dermatol. 2006;142:704-708.
- Livingston G, Brown A. Intermarriage in the U.S. 50 years after Loving v. Virginia. Pew Research Center website. https://www.pewsocialtrends.org/2017/05/18/intermarriage-in-the-u-s-50-years-after-loving-v-virginia/. Published May 18, 2017. Accessed April 15, 2020.
- He SY, McCulloch CE, Boscardin WJ, et al. Self-reported pigmentary phenotypes and race are significant but incomplete predictors of Fitzpatrick skin phototype in an ethnically diverse population. J Am Acad Dermatol. 2014;71:731-737.
Authors' Response
While Ms. Cruzval-O'Reilly and Dr. Lugo-Somolinos highlight many important points on conducting meaningful research for the Hispanic community, they seem to have misunderstood the overall purpose of our commentary,1 which was to highlight the increased skin cancer awareness that a notable and vocal member of the Hispanic community brought to our academic dermatology clinic, rather than to discuss skin types within the Hispanic community. As the authors mentioned, the term Hispanic is a descriptor of ethnicity rather than race, and Hispanic patients may have varying levels of skin pigmentation and sun sensitivity. While Dayanara Torres may have risk factors for developing melanoma, minimizing her connection with the Hispanic community because of her fair skin and light eyes would be a mistake. It not only isolates members of the Hispanic community that are of skin types I and II, but it also discounts the power of her story and language in raising awareness. We observed an increase in Hispanic patients presenting to our clinic who were concerned about skin cancer after Ms. Torres shared her diagnosis of metastatic melanoma through social media, followed by Spanish language educational videos on melanoma.
Several studies have described disparities among Hispanic patients diagnosed with melanoma as compared to their non-Hispanic white counterparts, including younger age at diagnosis, later stage of presentation, increased presence of regional involvement, and worse mortality.2-6 Furthermore, a small study of high school students by Ma et al7 showed disparities in skin cancer knowledge, perceived risk, and sun-protective behaviors among Hispanic whites and non-Hispanic whites, which remained significant (P<.05) after controlling for skin pigmentation and sun sensitivity. We agree with the authors that further analysis of skin type, race, genetics, and other risk factors may help refine the research on skin cancer disparities within the Hispanic community. We suspect that disparities may persist even when examining these factors. There have been several studies showing that knowledge-based interventions, especially when delivered in Spanish, improve understanding of skin cancer, personal risk, and self-examinations, and we support Ms. Torres' efforts in utilizing her platform to provide information about melanoma in Spanish.8-12
Radhika Srivastava, MD; Cindy Wassef, MD; Babar K. Rao, MD
From the Department of Dermatology, Rutgers Robert Wood Johnson Medical School, Somerset, New Jersey.
The authors report no conflict of interest.
Correspondence: Radhika Srivastava, MD, 1 World's Fair Dr, Ste 2400, Somerset, NJ 08873 ([email protected]).
References
- Srivastava R, Wassef C, Rao BK. The Dayanara effect: increasing skin cancer awareness in the Hispanic community. Cutis. 2019;103:257-258.
- Perez MI. Skin cancer in Hispanics in the United States. J Drugs Dermatol. 2019;18:s117-s120.
- Higgins S, Nazemi A, Feinstein S, et al. Clinical presentations of melanoma in African Americans, Hispanics, and Asians. Dermatol Surg. 2019;45:791-801.
- Harvey VM, Oldfield CW, Chen JT, et al. Melanoma disparities among US Hispanics: use of the social ecological model to contextualize reasons for inequitable outcomes and frame a research agenda [published online August 29, 2016]. J Skin Cancer. doi:10.1155/2016/4635740
- Garnett E, Townsend J, Steele B, et al. Characteristics, rates, and trends of melanoma incidence among Hispanics in the USA. Cancer Causes Control. 2016;27:647-659.
- Rouhani P, Hu S, Kirsner RS. Melanoma in Hispanic and black Americans. Cancer Control. 2008;15:248-253.
- Ma F, Collado-Mesa F, Hu S, et al. Skin cancer awareness and sun protection behaviors in white Hispanic and white non-Hispanic high school students in Miami, Florida. Arch Dermatol. 2007;143:983-988.
- Kundu RV, Kamaria M, Ortiz S, et al. Effectiveness of a knowledge-based intervention for melanoma among those with ethnic skin. J Am Acad Dermatol. 2010;62:777-784.
- Kailas A, Botwin AL, Pritchett EN, et al. Assessing the effectiveness of knowledge-based interventions in increasing skin cancer awareness, knowledge, and protective behaviors in skin of color populations. Cutis. 2017;100:235-240.
- Roman CJ, Guan X, Barnholtz-Sloan J, et al. A trial online educational melanoma program aimed at the Hispanic population improves knowledge and behaviors. Dermatol Surg. 2016;42:672-676.
- Hernandez C, Kim H, Mauleon G, et al. A pilot program in collaboration with community centers to increase awareness and participation in skin cancer screening among Latinos in Chicago. J Cancer Educ. 2013;28:342-345.
- Chung GY, Brown G, Gibson D. Increasing melanoma screening among Hispanic/Latino Americans: a community-based educational intervention. Health Educ Behav. 2015;42:627-632.
How aging affects melanoma development and treatment response
There are several mechanisms by which the aging microenvironment can drive cancer and influence response to therapy, according to a plenary presentation at the AACR virtual meeting II.
Ashani T. Weeraratna, PhD, highlighted research showing how the aging microenvironment affects tumor cell metabolism, angiogenesis, and treatment resistance in melanoma.
Dr. Weeraratna, of Johns Hopkins Bloomberg School of Public Health in Baltimore, first described a study showing how fibroblasts in the aged microenvironment contribute to tumor progression in models of melanoma (Nature. 2016 Apr 14;532[7598]:250-4).
Dr. Weeraratna and colleagues isolated dermal fibroblasts from young human donors (aged 25-35 years) and older donors (55-65 years) and used these cells to produce artificial skin.
Melanoma cells placed in the artificial skin created with young fibroblasts remained “very tightly nested at the surface,” Dr. Weeraratna said. On the other hand, melanoma cells migrated “very rapidly” through the artificial dermis created from aged fibroblasts.
In mouse models of melanoma, tumors grew much faster in young mice (6-8 weeks) than in old mice (12-18 months). However, tumors metastasized to the lung at a “much greater rate in the aged mice than in the young mice,” Dr. Weeraratna said.
Angiogenesis, SFRP2, and VEGF
Dr. Weeraratna went on to explain how a member of her lab conducted proteomic analyses of young and aged lung fibroblasts. The results were compared with results from prior analyses of young and aged skin fibroblasts.
The results showed that aged skin fibroblasts promote noncanonical WNT signaling via expression of SFRP2, SERPINE2, DKK1, Wnt5A, and ROR2. On the other hand, aged lung fibroblasts promote canonical WNT signaling via some of the same family members, including SFRP1, DKK3, and ROR1.
Research by another group showed that SFRP2 stimulates angiogenesis via a calcineurin/NFAT signaling pathway (Cancer Res. 2009 Jun 1;69[11]:4621-8).
Research in Dr. Weeraratna’s lab showed that SFRP2 and VEGF are inversely correlated with aging. Tumors in aged mice had an abundance of SFRP2 but little VEGF. Tumors in young mice had an abundance of VEGF but little SFRP2.
Dr. Weeraratna’s team wanted to determine if results would be similar in melanoma patients, so the researchers analyzed data from the TCGA database. They found that VEGF and two of its key receptors are decreased in older melanoma patients, in comparison with younger melanoma patients.
The clinical relevancy of this finding is reflected in an analysis of data from the AVAST-M study (Ann Oncol. 2019;30[12]:2013-4). When compared with observation, bevacizumab did not improve survival overall or for older patients, but the EGFR inhibitor was associated with longer survival in patients younger than 45 years.
Dr. Weeraratna said this finding and her group’s prior findings suggest younger melanoma patients have more VEGF but less angiogenesis than older patients. The older patients have less VEGF and more SFRP2, which drives angiogenesis.
Dr. Weeraratna’s lab then conducted experiments in young mice, which suggested that an anti-VEGF antibody can reduce angiogenesis, but not in the presence of SFRP2.
Lipid metabolism and treatment resistance
A recently published study by Dr. Weeraratna and colleagues tied changes in aged fibroblast lipid metabolism to treatment resistance in melanoma (Cancer Discov. 2020 Jun 4;CD-20-0329).
The research showed that melanoma cells accumulate lipids when incubated with aged, rather than young, fibroblasts in vitro.
Lipid uptake is mediated by fatty acid transporters (FATPs), and the researchers found that most FATPs were unchanged by age. However, FATP2 was elevated in melanoma cells exposed to aged media, aged mice, and melanoma patients older than 50 years of age.
When melanoma cells were incubated with conditioned media from aged fibroblasts and a FATP2 inhibitor, they no longer took up lipids.
When FATP2 was knocked down in aged mice with melanoma, BRAF and MEK inhibitors (which are not very effective ordinarily) caused dramatic and prolonged tumor regression. These effects were not seen with FATP2 inhibition in young mice.
These results suggest FATP2 is a key transporter of lipids in the aged microenvironment, and inhibiting FATP2 can delay the onset of treatment resistance.
Striving to understand a complex system
For many years, the dogma was that cancer cells behaved like unwelcome invaders, co-opting the metabolic machinery of the sites of spread, with crowding of the normal structures within those organs.
To say that concept was primitive is an understatement. Clearly, the relationship between tumor cells and the surrounding stroma is complex. Changes that occur in an aging microenvironment can influence cancer outcomes in older adults.
Dr. Weeraratna’s presentation adds further impetus to efforts to broaden eligibility criteria for clinical trials so the median age and the metabolic milieu of trial participants more closely parallels the general population.
She highlighted the importance of data analysis by age cohorts and the need to design preclinical studies so that investigators can study the microenvironment of cancer cells in in vitro models and in young and older laboratory animals.
As management expert W. Edwards Deming is believed to have said, “Every system is perfectly designed to get the results it gets.” Cancer is likely not an independent, hostile invader, overtaking the failing machinery of aging cells. To understand the intersection of cancer and aging, we need a more perfect understanding of the system in which tumors develop and are treated.
Dr. Weeraratna reported having no disclosures.
Dr. Lyss was a community-based medical oncologist and clinical researcher for more than 35 years before his recent retirement. His clinical and research interests were focused on breast and lung cancers as well as expanding clinical trial access to medically underserved populations. He is based in St. Louis. He has no conflicts of interest.
SOURCE: Weeraratna A. AACR 2020. Age against the machine: How the aging microenvironment governs response to therapy.
There are several mechanisms by which the aging microenvironment can drive cancer and influence response to therapy, according to a plenary presentation at the AACR virtual meeting II.
Ashani T. Weeraratna, PhD, highlighted research showing how the aging microenvironment affects tumor cell metabolism, angiogenesis, and treatment resistance in melanoma.
Dr. Weeraratna, of Johns Hopkins Bloomberg School of Public Health in Baltimore, first described a study showing how fibroblasts in the aged microenvironment contribute to tumor progression in models of melanoma (Nature. 2016 Apr 14;532[7598]:250-4).
Dr. Weeraratna and colleagues isolated dermal fibroblasts from young human donors (aged 25-35 years) and older donors (55-65 years) and used these cells to produce artificial skin.
Melanoma cells placed in the artificial skin created with young fibroblasts remained “very tightly nested at the surface,” Dr. Weeraratna said. On the other hand, melanoma cells migrated “very rapidly” through the artificial dermis created from aged fibroblasts.
In mouse models of melanoma, tumors grew much faster in young mice (6-8 weeks) than in old mice (12-18 months). However, tumors metastasized to the lung at a “much greater rate in the aged mice than in the young mice,” Dr. Weeraratna said.
Angiogenesis, SFRP2, and VEGF
Dr. Weeraratna went on to explain how a member of her lab conducted proteomic analyses of young and aged lung fibroblasts. The results were compared with results from prior analyses of young and aged skin fibroblasts.
The results showed that aged skin fibroblasts promote noncanonical WNT signaling via expression of SFRP2, SERPINE2, DKK1, Wnt5A, and ROR2. On the other hand, aged lung fibroblasts promote canonical WNT signaling via some of the same family members, including SFRP1, DKK3, and ROR1.
Research by another group showed that SFRP2 stimulates angiogenesis via a calcineurin/NFAT signaling pathway (Cancer Res. 2009 Jun 1;69[11]:4621-8).
Research in Dr. Weeraratna’s lab showed that SFRP2 and VEGF are inversely correlated with aging. Tumors in aged mice had an abundance of SFRP2 but little VEGF. Tumors in young mice had an abundance of VEGF but little SFRP2.
Dr. Weeraratna’s team wanted to determine if results would be similar in melanoma patients, so the researchers analyzed data from the TCGA database. They found that VEGF and two of its key receptors are decreased in older melanoma patients, in comparison with younger melanoma patients.
The clinical relevancy of this finding is reflected in an analysis of data from the AVAST-M study (Ann Oncol. 2019;30[12]:2013-4). When compared with observation, bevacizumab did not improve survival overall or for older patients, but the EGFR inhibitor was associated with longer survival in patients younger than 45 years.
Dr. Weeraratna said this finding and her group’s prior findings suggest younger melanoma patients have more VEGF but less angiogenesis than older patients. The older patients have less VEGF and more SFRP2, which drives angiogenesis.
Dr. Weeraratna’s lab then conducted experiments in young mice, which suggested that an anti-VEGF antibody can reduce angiogenesis, but not in the presence of SFRP2.
Lipid metabolism and treatment resistance
A recently published study by Dr. Weeraratna and colleagues tied changes in aged fibroblast lipid metabolism to treatment resistance in melanoma (Cancer Discov. 2020 Jun 4;CD-20-0329).
The research showed that melanoma cells accumulate lipids when incubated with aged, rather than young, fibroblasts in vitro.
Lipid uptake is mediated by fatty acid transporters (FATPs), and the researchers found that most FATPs were unchanged by age. However, FATP2 was elevated in melanoma cells exposed to aged media, aged mice, and melanoma patients older than 50 years of age.
When melanoma cells were incubated with conditioned media from aged fibroblasts and a FATP2 inhibitor, they no longer took up lipids.
When FATP2 was knocked down in aged mice with melanoma, BRAF and MEK inhibitors (which are not very effective ordinarily) caused dramatic and prolonged tumor regression. These effects were not seen with FATP2 inhibition in young mice.
These results suggest FATP2 is a key transporter of lipids in the aged microenvironment, and inhibiting FATP2 can delay the onset of treatment resistance.
Striving to understand a complex system
For many years, the dogma was that cancer cells behaved like unwelcome invaders, co-opting the metabolic machinery of the sites of spread, with crowding of the normal structures within those organs.
To say that concept was primitive is an understatement. Clearly, the relationship between tumor cells and the surrounding stroma is complex. Changes that occur in an aging microenvironment can influence cancer outcomes in older adults.
Dr. Weeraratna’s presentation adds further impetus to efforts to broaden eligibility criteria for clinical trials so the median age and the metabolic milieu of trial participants more closely parallels the general population.
She highlighted the importance of data analysis by age cohorts and the need to design preclinical studies so that investigators can study the microenvironment of cancer cells in in vitro models and in young and older laboratory animals.
As management expert W. Edwards Deming is believed to have said, “Every system is perfectly designed to get the results it gets.” Cancer is likely not an independent, hostile invader, overtaking the failing machinery of aging cells. To understand the intersection of cancer and aging, we need a more perfect understanding of the system in which tumors develop and are treated.
Dr. Weeraratna reported having no disclosures.
Dr. Lyss was a community-based medical oncologist and clinical researcher for more than 35 years before his recent retirement. His clinical and research interests were focused on breast and lung cancers as well as expanding clinical trial access to medically underserved populations. He is based in St. Louis. He has no conflicts of interest.
SOURCE: Weeraratna A. AACR 2020. Age against the machine: How the aging microenvironment governs response to therapy.
There are several mechanisms by which the aging microenvironment can drive cancer and influence response to therapy, according to a plenary presentation at the AACR virtual meeting II.
Ashani T. Weeraratna, PhD, highlighted research showing how the aging microenvironment affects tumor cell metabolism, angiogenesis, and treatment resistance in melanoma.
Dr. Weeraratna, of Johns Hopkins Bloomberg School of Public Health in Baltimore, first described a study showing how fibroblasts in the aged microenvironment contribute to tumor progression in models of melanoma (Nature. 2016 Apr 14;532[7598]:250-4).
Dr. Weeraratna and colleagues isolated dermal fibroblasts from young human donors (aged 25-35 years) and older donors (55-65 years) and used these cells to produce artificial skin.
Melanoma cells placed in the artificial skin created with young fibroblasts remained “very tightly nested at the surface,” Dr. Weeraratna said. On the other hand, melanoma cells migrated “very rapidly” through the artificial dermis created from aged fibroblasts.
In mouse models of melanoma, tumors grew much faster in young mice (6-8 weeks) than in old mice (12-18 months). However, tumors metastasized to the lung at a “much greater rate in the aged mice than in the young mice,” Dr. Weeraratna said.
Angiogenesis, SFRP2, and VEGF
Dr. Weeraratna went on to explain how a member of her lab conducted proteomic analyses of young and aged lung fibroblasts. The results were compared with results from prior analyses of young and aged skin fibroblasts.
The results showed that aged skin fibroblasts promote noncanonical WNT signaling via expression of SFRP2, SERPINE2, DKK1, Wnt5A, and ROR2. On the other hand, aged lung fibroblasts promote canonical WNT signaling via some of the same family members, including SFRP1, DKK3, and ROR1.
Research by another group showed that SFRP2 stimulates angiogenesis via a calcineurin/NFAT signaling pathway (Cancer Res. 2009 Jun 1;69[11]:4621-8).
Research in Dr. Weeraratna’s lab showed that SFRP2 and VEGF are inversely correlated with aging. Tumors in aged mice had an abundance of SFRP2 but little VEGF. Tumors in young mice had an abundance of VEGF but little SFRP2.
Dr. Weeraratna’s team wanted to determine if results would be similar in melanoma patients, so the researchers analyzed data from the TCGA database. They found that VEGF and two of its key receptors are decreased in older melanoma patients, in comparison with younger melanoma patients.
The clinical relevancy of this finding is reflected in an analysis of data from the AVAST-M study (Ann Oncol. 2019;30[12]:2013-4). When compared with observation, bevacizumab did not improve survival overall or for older patients, but the EGFR inhibitor was associated with longer survival in patients younger than 45 years.
Dr. Weeraratna said this finding and her group’s prior findings suggest younger melanoma patients have more VEGF but less angiogenesis than older patients. The older patients have less VEGF and more SFRP2, which drives angiogenesis.
Dr. Weeraratna’s lab then conducted experiments in young mice, which suggested that an anti-VEGF antibody can reduce angiogenesis, but not in the presence of SFRP2.
Lipid metabolism and treatment resistance
A recently published study by Dr. Weeraratna and colleagues tied changes in aged fibroblast lipid metabolism to treatment resistance in melanoma (Cancer Discov. 2020 Jun 4;CD-20-0329).
The research showed that melanoma cells accumulate lipids when incubated with aged, rather than young, fibroblasts in vitro.
Lipid uptake is mediated by fatty acid transporters (FATPs), and the researchers found that most FATPs were unchanged by age. However, FATP2 was elevated in melanoma cells exposed to aged media, aged mice, and melanoma patients older than 50 years of age.
When melanoma cells were incubated with conditioned media from aged fibroblasts and a FATP2 inhibitor, they no longer took up lipids.
When FATP2 was knocked down in aged mice with melanoma, BRAF and MEK inhibitors (which are not very effective ordinarily) caused dramatic and prolonged tumor regression. These effects were not seen with FATP2 inhibition in young mice.
These results suggest FATP2 is a key transporter of lipids in the aged microenvironment, and inhibiting FATP2 can delay the onset of treatment resistance.
Striving to understand a complex system
For many years, the dogma was that cancer cells behaved like unwelcome invaders, co-opting the metabolic machinery of the sites of spread, with crowding of the normal structures within those organs.
To say that concept was primitive is an understatement. Clearly, the relationship between tumor cells and the surrounding stroma is complex. Changes that occur in an aging microenvironment can influence cancer outcomes in older adults.
Dr. Weeraratna’s presentation adds further impetus to efforts to broaden eligibility criteria for clinical trials so the median age and the metabolic milieu of trial participants more closely parallels the general population.
She highlighted the importance of data analysis by age cohorts and the need to design preclinical studies so that investigators can study the microenvironment of cancer cells in in vitro models and in young and older laboratory animals.
As management expert W. Edwards Deming is believed to have said, “Every system is perfectly designed to get the results it gets.” Cancer is likely not an independent, hostile invader, overtaking the failing machinery of aging cells. To understand the intersection of cancer and aging, we need a more perfect understanding of the system in which tumors develop and are treated.
Dr. Weeraratna reported having no disclosures.
Dr. Lyss was a community-based medical oncologist and clinical researcher for more than 35 years before his recent retirement. His clinical and research interests were focused on breast and lung cancers as well as expanding clinical trial access to medically underserved populations. He is based in St. Louis. He has no conflicts of interest.
SOURCE: Weeraratna A. AACR 2020. Age against the machine: How the aging microenvironment governs response to therapy.
FROM AACR 2020
Cutaneous Metastases Masquerading as Solitary or Multiple Keratoacanthomas
To the Editor:
We read with interest the excellent Cutis articles on cutaneous metastases by Tarantino et al1 and Agnetta et al.2 Tarantino et al1 reported a 59-year-old man who developed cutaneous metastases on the scalp from an esophageal adenocarcinoma. Agnetta et al2 described a 76-year-old woman with metastatic melanoma mimicking eruptive keratoacanthomas.
Cutaneous metastases are not common. They may herald the unsuspected diagnosis of a solid tumor recurrence or progression of systemic disease in an oncology patient. Occasionally, they are the primary manifestation of a visceral tumor in a previously cancer-free patient. Less often, skin lesions are the manifestation of a new or recurrent hematologic malignancy.3,4
The morphology of cutaneous metastases is variable. Most commonly they appear as papules and nodules. However, they can mimic bacterial (eg, erysipelas) and viral (eg, herpes zoster) infections or present as scalp alopecia.5-7
Cutaneous metastases also can mimic benign (eg, epidermoid cysts) or malignant (eg, keratoacanthoma) neoplasms. Keratoacanthomalike cutaneous metastases are rare.8 They can present as single or multiple tumors.9,10
In the case reported by Tarantino et al,1 the patient had a history of metastatic adenocarcinoma of the esophagus. His unsuspected recurrence presented not only with a single keratoacanthomalike cutaneous metastasis on the scalp but also with another metastasis-related scalp lesion that appeared as a smooth pearly papule. We also observed a 53-year-old man whose metastatic esophageal adenocarcinoma presented with a keratoacanthomalike nodule on the right upper lip; additionally, the patient had other cutaneous metastases that appeared as an erythematous papule on the forehead and a cystic nodule on the scalp.8 Other investigators also observed a single keratoacanthomalike lesion on the left cheek of a 49-year-old man with metastatic esophageal adenocarcinoma.11
Agnetta et al2 described a patient with a history of malignant melanoma on the left upper back that had been excised 2 years prior. She presented with the eruptive onset of multiple keratoacanthomalike cutaneous metastases on the chest, back, and right arm.2 The important observation of metastatic malignant melanoma presenting as multiple keratoacanthomalike cutaneous metastases pointed out by Agnetta et al2 confirms a similar occurrence reported by Reed et al12 in a patient with metastatic malignant melanoma.
We also previously reported the case of a 68-year-old man with metastatic laryngeal squamous cell carcinoma (SCC) who developed more than 10 keratoacanthomalike nodules within a radiation port that extended from the face to the mid chest.10 In addition, other researchers have noted a similar phenomenon of keratoacanthomalike cutaneous metastases mimicking eruptive keratoacanthomas.13 Gil et al14 described a 40-year-old woman whose metastatic epithelioid trophoblastic tumor initially presented as 11 keratoacanthomalike scalp nodules; interestingly, the first nodule spontaneously regressed. Araghi et al15 reported a 58-year-old woman--with a stable SCC of the larynx that had been diagnosed 2 years prior and treated with chemoradiotherapy--in whom cancer progression presented as multiple keratoacanthomalike lesions in an area of prior radiotherapy.
In conclusion, cutaneous metastases presenting as new-onset solitary or multiple keratoacanthomalike nodules in either a cancer-free individual or a patient with a prior history of a visceral malignancy is uncommon. Although the clinical features mimic those of a single or eruptive keratoacanthomas, a biopsy will readily establish the diagnosis of cutaneous metastatic cancer. Metastatic esophageal carcinoma--either adenocarcinoma or SCC--can present, albeit rarely, with cutaneous lesions that can have various morphologies.8 Whether there is an increased predilection for patients with metastatic esophageal adenocarcinoma to present with single keratoacanthomalike cutaneous metastases with or without concurrent additional skin lesions of cutaneous metastases of other morphologies remains to be determined.
- Tarantino IS, Tennill T, Fraga G, et al. Cutaneous metastases from esophageal adenocarcinoma on the scalp. Cutis. 2020;105:E3-E5.
- Agnetta V, Hamstra A, Hirokane J, et al. Metastatic melanoma mimicking eruptive keratoacanthomas. Cutis. 2020;105:E29-E31.
- Cohen PR. Skin clues to primary and metastatic malignancy. Am Fam Physician. 1995;51:1199-1204.
- Cohen PR. Leukemia cutis-associated leonine facies and eyebrow loss. Cutis. 2019;103:212.
- Cohen PR, Prieto VG, Piha-Paul SA, et al. The "shield sign" in two men with metastatic salivary duct carcinoma to the skin: cutaneous metastases presenting as carcinoma hemorrhagiectoides. J Clin Aesthet Dermatol. 2012;5:27-36.
- Manteaux A, Cohen PR, Rapini RP. Zosteriform and epidermotropic metastasis. report of two cases. J Dermatol Surg Oncol. 1992;18:97-100.
- Conner KB, Cohen PR. Cutaneous metastases of breast carcinoma presenting as alopecia neoplastica. South Med J. 2009;102:385-389.
- Riahi RR, Cohen PR. Clinical manifestations of cutaneous metastases: a review with special emphasis on cutaneous metastases mimicking keratoacanthoma. Am J Clin Dermatol. 2012;13:103-112.
- Riahi RR, Cohen PR. Malignancies with skin lesions mimicking keratoacanthoma. Dermatol Online J. 2013;19:20397.
- Ellis DL, Riahi RR, Murina AT, et al. Metastatic laryngeal carcinoma mimicking eruptive keratoacanthomas: report of keratoacanthoma-like cutaneous metastases in a radiation port. Dermatol Online J. 2014;20. pii:13030/qt3s43b81f.
- Hani AC, Nuñez E, Cuellar I, et al. Cutaneous metastases as a manifestation of esophageal adenocarcinoma recurrence: a case report [published online September 5, 2019]. Rev Gastroenterol Mex. doi:10.1016/j.rgmx.2019.06.002.
- Reed KB, Cook-Norris RH, Brewer JD. The cutaneous manifestations of metastatic malignant melanoma. Int J Dermatol. 2012;51:243-249.
- Cohen PR, Riahi RR. Cutaneous metastases mimicking keratoacanthoma. Int J Dermatol. 2014;53:E320-E322.
- Gil F, Elvas L, Raposo S, et al. Keratoacanthoma-like nodules as first manifestation of metastatic epithelioid trophoblastic tumor. Dermatol Online J. 2019;25. pii:13030/qt9xx6p2tt.
- Araghi F, Fatemi A, Rakhshan A, et al. Skin metastasis of laryngeal carcinoma presenting as multiple eruptive nodules [published online February 10, 2020]. Head Neck Pathol. doi:10.1007/s12105-020-01143-1.
To the Editor:
We read with interest the excellent Cutis articles on cutaneous metastases by Tarantino et al1 and Agnetta et al.2 Tarantino et al1 reported a 59-year-old man who developed cutaneous metastases on the scalp from an esophageal adenocarcinoma. Agnetta et al2 described a 76-year-old woman with metastatic melanoma mimicking eruptive keratoacanthomas.
Cutaneous metastases are not common. They may herald the unsuspected diagnosis of a solid tumor recurrence or progression of systemic disease in an oncology patient. Occasionally, they are the primary manifestation of a visceral tumor in a previously cancer-free patient. Less often, skin lesions are the manifestation of a new or recurrent hematologic malignancy.3,4
The morphology of cutaneous metastases is variable. Most commonly they appear as papules and nodules. However, they can mimic bacterial (eg, erysipelas) and viral (eg, herpes zoster) infections or present as scalp alopecia.5-7
Cutaneous metastases also can mimic benign (eg, epidermoid cysts) or malignant (eg, keratoacanthoma) neoplasms. Keratoacanthomalike cutaneous metastases are rare.8 They can present as single or multiple tumors.9,10
In the case reported by Tarantino et al,1 the patient had a history of metastatic adenocarcinoma of the esophagus. His unsuspected recurrence presented not only with a single keratoacanthomalike cutaneous metastasis on the scalp but also with another metastasis-related scalp lesion that appeared as a smooth pearly papule. We also observed a 53-year-old man whose metastatic esophageal adenocarcinoma presented with a keratoacanthomalike nodule on the right upper lip; additionally, the patient had other cutaneous metastases that appeared as an erythematous papule on the forehead and a cystic nodule on the scalp.8 Other investigators also observed a single keratoacanthomalike lesion on the left cheek of a 49-year-old man with metastatic esophageal adenocarcinoma.11
Agnetta et al2 described a patient with a history of malignant melanoma on the left upper back that had been excised 2 years prior. She presented with the eruptive onset of multiple keratoacanthomalike cutaneous metastases on the chest, back, and right arm.2 The important observation of metastatic malignant melanoma presenting as multiple keratoacanthomalike cutaneous metastases pointed out by Agnetta et al2 confirms a similar occurrence reported by Reed et al12 in a patient with metastatic malignant melanoma.
We also previously reported the case of a 68-year-old man with metastatic laryngeal squamous cell carcinoma (SCC) who developed more than 10 keratoacanthomalike nodules within a radiation port that extended from the face to the mid chest.10 In addition, other researchers have noted a similar phenomenon of keratoacanthomalike cutaneous metastases mimicking eruptive keratoacanthomas.13 Gil et al14 described a 40-year-old woman whose metastatic epithelioid trophoblastic tumor initially presented as 11 keratoacanthomalike scalp nodules; interestingly, the first nodule spontaneously regressed. Araghi et al15 reported a 58-year-old woman--with a stable SCC of the larynx that had been diagnosed 2 years prior and treated with chemoradiotherapy--in whom cancer progression presented as multiple keratoacanthomalike lesions in an area of prior radiotherapy.
In conclusion, cutaneous metastases presenting as new-onset solitary or multiple keratoacanthomalike nodules in either a cancer-free individual or a patient with a prior history of a visceral malignancy is uncommon. Although the clinical features mimic those of a single or eruptive keratoacanthomas, a biopsy will readily establish the diagnosis of cutaneous metastatic cancer. Metastatic esophageal carcinoma--either adenocarcinoma or SCC--can present, albeit rarely, with cutaneous lesions that can have various morphologies.8 Whether there is an increased predilection for patients with metastatic esophageal adenocarcinoma to present with single keratoacanthomalike cutaneous metastases with or without concurrent additional skin lesions of cutaneous metastases of other morphologies remains to be determined.
To the Editor:
We read with interest the excellent Cutis articles on cutaneous metastases by Tarantino et al1 and Agnetta et al.2 Tarantino et al1 reported a 59-year-old man who developed cutaneous metastases on the scalp from an esophageal adenocarcinoma. Agnetta et al2 described a 76-year-old woman with metastatic melanoma mimicking eruptive keratoacanthomas.
Cutaneous metastases are not common. They may herald the unsuspected diagnosis of a solid tumor recurrence or progression of systemic disease in an oncology patient. Occasionally, they are the primary manifestation of a visceral tumor in a previously cancer-free patient. Less often, skin lesions are the manifestation of a new or recurrent hematologic malignancy.3,4
The morphology of cutaneous metastases is variable. Most commonly they appear as papules and nodules. However, they can mimic bacterial (eg, erysipelas) and viral (eg, herpes zoster) infections or present as scalp alopecia.5-7
Cutaneous metastases also can mimic benign (eg, epidermoid cysts) or malignant (eg, keratoacanthoma) neoplasms. Keratoacanthomalike cutaneous metastases are rare.8 They can present as single or multiple tumors.9,10
In the case reported by Tarantino et al,1 the patient had a history of metastatic adenocarcinoma of the esophagus. His unsuspected recurrence presented not only with a single keratoacanthomalike cutaneous metastasis on the scalp but also with another metastasis-related scalp lesion that appeared as a smooth pearly papule. We also observed a 53-year-old man whose metastatic esophageal adenocarcinoma presented with a keratoacanthomalike nodule on the right upper lip; additionally, the patient had other cutaneous metastases that appeared as an erythematous papule on the forehead and a cystic nodule on the scalp.8 Other investigators also observed a single keratoacanthomalike lesion on the left cheek of a 49-year-old man with metastatic esophageal adenocarcinoma.11
Agnetta et al2 described a patient with a history of malignant melanoma on the left upper back that had been excised 2 years prior. She presented with the eruptive onset of multiple keratoacanthomalike cutaneous metastases on the chest, back, and right arm.2 The important observation of metastatic malignant melanoma presenting as multiple keratoacanthomalike cutaneous metastases pointed out by Agnetta et al2 confirms a similar occurrence reported by Reed et al12 in a patient with metastatic malignant melanoma.
We also previously reported the case of a 68-year-old man with metastatic laryngeal squamous cell carcinoma (SCC) who developed more than 10 keratoacanthomalike nodules within a radiation port that extended from the face to the mid chest.10 In addition, other researchers have noted a similar phenomenon of keratoacanthomalike cutaneous metastases mimicking eruptive keratoacanthomas.13 Gil et al14 described a 40-year-old woman whose metastatic epithelioid trophoblastic tumor initially presented as 11 keratoacanthomalike scalp nodules; interestingly, the first nodule spontaneously regressed. Araghi et al15 reported a 58-year-old woman--with a stable SCC of the larynx that had been diagnosed 2 years prior and treated with chemoradiotherapy--in whom cancer progression presented as multiple keratoacanthomalike lesions in an area of prior radiotherapy.
In conclusion, cutaneous metastases presenting as new-onset solitary or multiple keratoacanthomalike nodules in either a cancer-free individual or a patient with a prior history of a visceral malignancy is uncommon. Although the clinical features mimic those of a single or eruptive keratoacanthomas, a biopsy will readily establish the diagnosis of cutaneous metastatic cancer. Metastatic esophageal carcinoma--either adenocarcinoma or SCC--can present, albeit rarely, with cutaneous lesions that can have various morphologies.8 Whether there is an increased predilection for patients with metastatic esophageal adenocarcinoma to present with single keratoacanthomalike cutaneous metastases with or without concurrent additional skin lesions of cutaneous metastases of other morphologies remains to be determined.
- Tarantino IS, Tennill T, Fraga G, et al. Cutaneous metastases from esophageal adenocarcinoma on the scalp. Cutis. 2020;105:E3-E5.
- Agnetta V, Hamstra A, Hirokane J, et al. Metastatic melanoma mimicking eruptive keratoacanthomas. Cutis. 2020;105:E29-E31.
- Cohen PR. Skin clues to primary and metastatic malignancy. Am Fam Physician. 1995;51:1199-1204.
- Cohen PR. Leukemia cutis-associated leonine facies and eyebrow loss. Cutis. 2019;103:212.
- Cohen PR, Prieto VG, Piha-Paul SA, et al. The "shield sign" in two men with metastatic salivary duct carcinoma to the skin: cutaneous metastases presenting as carcinoma hemorrhagiectoides. J Clin Aesthet Dermatol. 2012;5:27-36.
- Manteaux A, Cohen PR, Rapini RP. Zosteriform and epidermotropic metastasis. report of two cases. J Dermatol Surg Oncol. 1992;18:97-100.
- Conner KB, Cohen PR. Cutaneous metastases of breast carcinoma presenting as alopecia neoplastica. South Med J. 2009;102:385-389.
- Riahi RR, Cohen PR. Clinical manifestations of cutaneous metastases: a review with special emphasis on cutaneous metastases mimicking keratoacanthoma. Am J Clin Dermatol. 2012;13:103-112.
- Riahi RR, Cohen PR. Malignancies with skin lesions mimicking keratoacanthoma. Dermatol Online J. 2013;19:20397.
- Ellis DL, Riahi RR, Murina AT, et al. Metastatic laryngeal carcinoma mimicking eruptive keratoacanthomas: report of keratoacanthoma-like cutaneous metastases in a radiation port. Dermatol Online J. 2014;20. pii:13030/qt3s43b81f.
- Hani AC, Nuñez E, Cuellar I, et al. Cutaneous metastases as a manifestation of esophageal adenocarcinoma recurrence: a case report [published online September 5, 2019]. Rev Gastroenterol Mex. doi:10.1016/j.rgmx.2019.06.002.
- Reed KB, Cook-Norris RH, Brewer JD. The cutaneous manifestations of metastatic malignant melanoma. Int J Dermatol. 2012;51:243-249.
- Cohen PR, Riahi RR. Cutaneous metastases mimicking keratoacanthoma. Int J Dermatol. 2014;53:E320-E322.
- Gil F, Elvas L, Raposo S, et al. Keratoacanthoma-like nodules as first manifestation of metastatic epithelioid trophoblastic tumor. Dermatol Online J. 2019;25. pii:13030/qt9xx6p2tt.
- Araghi F, Fatemi A, Rakhshan A, et al. Skin metastasis of laryngeal carcinoma presenting as multiple eruptive nodules [published online February 10, 2020]. Head Neck Pathol. doi:10.1007/s12105-020-01143-1.
- Tarantino IS, Tennill T, Fraga G, et al. Cutaneous metastases from esophageal adenocarcinoma on the scalp. Cutis. 2020;105:E3-E5.
- Agnetta V, Hamstra A, Hirokane J, et al. Metastatic melanoma mimicking eruptive keratoacanthomas. Cutis. 2020;105:E29-E31.
- Cohen PR. Skin clues to primary and metastatic malignancy. Am Fam Physician. 1995;51:1199-1204.
- Cohen PR. Leukemia cutis-associated leonine facies and eyebrow loss. Cutis. 2019;103:212.
- Cohen PR, Prieto VG, Piha-Paul SA, et al. The "shield sign" in two men with metastatic salivary duct carcinoma to the skin: cutaneous metastases presenting as carcinoma hemorrhagiectoides. J Clin Aesthet Dermatol. 2012;5:27-36.
- Manteaux A, Cohen PR, Rapini RP. Zosteriform and epidermotropic metastasis. report of two cases. J Dermatol Surg Oncol. 1992;18:97-100.
- Conner KB, Cohen PR. Cutaneous metastases of breast carcinoma presenting as alopecia neoplastica. South Med J. 2009;102:385-389.
- Riahi RR, Cohen PR. Clinical manifestations of cutaneous metastases: a review with special emphasis on cutaneous metastases mimicking keratoacanthoma. Am J Clin Dermatol. 2012;13:103-112.
- Riahi RR, Cohen PR. Malignancies with skin lesions mimicking keratoacanthoma. Dermatol Online J. 2013;19:20397.
- Ellis DL, Riahi RR, Murina AT, et al. Metastatic laryngeal carcinoma mimicking eruptive keratoacanthomas: report of keratoacanthoma-like cutaneous metastases in a radiation port. Dermatol Online J. 2014;20. pii:13030/qt3s43b81f.
- Hani AC, Nuñez E, Cuellar I, et al. Cutaneous metastases as a manifestation of esophageal adenocarcinoma recurrence: a case report [published online September 5, 2019]. Rev Gastroenterol Mex. doi:10.1016/j.rgmx.2019.06.002.
- Reed KB, Cook-Norris RH, Brewer JD. The cutaneous manifestations of metastatic malignant melanoma. Int J Dermatol. 2012;51:243-249.
- Cohen PR, Riahi RR. Cutaneous metastases mimicking keratoacanthoma. Int J Dermatol. 2014;53:E320-E322.
- Gil F, Elvas L, Raposo S, et al. Keratoacanthoma-like nodules as first manifestation of metastatic epithelioid trophoblastic tumor. Dermatol Online J. 2019;25. pii:13030/qt9xx6p2tt.
- Araghi F, Fatemi A, Rakhshan A, et al. Skin metastasis of laryngeal carcinoma presenting as multiple eruptive nodules [published online February 10, 2020]. Head Neck Pathol. doi:10.1007/s12105-020-01143-1.
Real-world data support adjuvant immunotherapy for stage III melanoma
Among patients with stage IIIC disease, the 2-year survival rate was 70% in those who received immunotherapy and 59% in those who did not (P < .01). The median overall survival in this group was 32.8 months with immunotherapy and 28 months without it (P < .01).
Among patients with stage IIIA disease, the 2-year survival rate was 94% with immunotherapy and 91% without it (P = .03).
There was a trend toward a 2-year survival benefit with immunotherapy in patients with stage IIIB disease and in all 4,094 stage III patients, but the differences were not significant. The 2-year survival rate was 84% with immunotherapy and 81% without it among patients with stage IIIB disease (P = .35). The survival rates were 83% and 80%, respectively, in all stage III patients (P = .051).
This was an early analysis, noted investigator Justin Moyers, MD, of Loma Linda (Calif.) University. Ipilimumab was approved as adjuvant therapy for stage III melanoma patients in 2015, the year patients from this analysis were diagnosed.
“There’s really only 2 full years of survival data,” Dr. Moyers said. “I think given time, we will see a benefit amongst all the substages.”
In the meantime, “I would definitely not use this data to say whether or not [immunotherapy] should be given,” Dr. Moyers said.
The researchers were just using the database – which captures 52% of U.S. melanoma cases – to see if “real-world data mimics the clinical trial data,” Dr. Moyers said.
Overall, the findings support “adjuvant immunotherapy in the real-world setting,” he said.
The researchers also looked at treatment patterns in 2015-2016 across 8,160 patients with stage III melanoma, 4,094 of whom were included in the aforementioned survival analysis. There were 2,260 patients (27.7%) who received immunotherapy after surgery during that time period.
Uptake of adjuvant immunotherapy “was low to start, but those patients did better than ones who did not get” it, said AACR president Antoni Ribas, MD, PhD, of the University of California, Los Angeles, who was not involved in the study.
Immunotherapy recipients were younger, on average (54.8 years vs. 62.4 years). Patients with Charlson comorbidity scores above zero and those on Medicare were less likely to receive immunotherapy (18.4% Medicare vs. over 30% with other payers). There also were trends of decreased use with lower income and lower high school graduation rates.
The finding “highlights the negative impact of socioeconomic [factors] on access to proven therapy,” Dr. Ribas said.
As for low use among Medicare patients, uptake of new treatments, in general, “seems to be faster with private insurance,” he noted.
The study excluded patients who received systemic therapies other than immunotherapy, as well as those who received immunotherapy before surgery. Among study limitations, the specific immunotherapies patients received was unknown.
There was no external funding for this study. Dr. Moyers reported travel compensation from Astellas Pharmaceuticals in 2018. Dr. Ribas disclosed relationships with Amgen, Chugai, Merck, Sanofi, Tango, Arcus, Bioncotech, Compugen, CytomX, FLX Bio, ImaginAb, Isoplexis, Merus, Rgenix, and PACT.
SOURCE: Moyers J et al. AACR 2020, Abstract 4338.
Among patients with stage IIIC disease, the 2-year survival rate was 70% in those who received immunotherapy and 59% in those who did not (P < .01). The median overall survival in this group was 32.8 months with immunotherapy and 28 months without it (P < .01).
Among patients with stage IIIA disease, the 2-year survival rate was 94% with immunotherapy and 91% without it (P = .03).
There was a trend toward a 2-year survival benefit with immunotherapy in patients with stage IIIB disease and in all 4,094 stage III patients, but the differences were not significant. The 2-year survival rate was 84% with immunotherapy and 81% without it among patients with stage IIIB disease (P = .35). The survival rates were 83% and 80%, respectively, in all stage III patients (P = .051).
This was an early analysis, noted investigator Justin Moyers, MD, of Loma Linda (Calif.) University. Ipilimumab was approved as adjuvant therapy for stage III melanoma patients in 2015, the year patients from this analysis were diagnosed.
“There’s really only 2 full years of survival data,” Dr. Moyers said. “I think given time, we will see a benefit amongst all the substages.”
In the meantime, “I would definitely not use this data to say whether or not [immunotherapy] should be given,” Dr. Moyers said.
The researchers were just using the database – which captures 52% of U.S. melanoma cases – to see if “real-world data mimics the clinical trial data,” Dr. Moyers said.
Overall, the findings support “adjuvant immunotherapy in the real-world setting,” he said.
The researchers also looked at treatment patterns in 2015-2016 across 8,160 patients with stage III melanoma, 4,094 of whom were included in the aforementioned survival analysis. There were 2,260 patients (27.7%) who received immunotherapy after surgery during that time period.
Uptake of adjuvant immunotherapy “was low to start, but those patients did better than ones who did not get” it, said AACR president Antoni Ribas, MD, PhD, of the University of California, Los Angeles, who was not involved in the study.
Immunotherapy recipients were younger, on average (54.8 years vs. 62.4 years). Patients with Charlson comorbidity scores above zero and those on Medicare were less likely to receive immunotherapy (18.4% Medicare vs. over 30% with other payers). There also were trends of decreased use with lower income and lower high school graduation rates.
The finding “highlights the negative impact of socioeconomic [factors] on access to proven therapy,” Dr. Ribas said.
As for low use among Medicare patients, uptake of new treatments, in general, “seems to be faster with private insurance,” he noted.
The study excluded patients who received systemic therapies other than immunotherapy, as well as those who received immunotherapy before surgery. Among study limitations, the specific immunotherapies patients received was unknown.
There was no external funding for this study. Dr. Moyers reported travel compensation from Astellas Pharmaceuticals in 2018. Dr. Ribas disclosed relationships with Amgen, Chugai, Merck, Sanofi, Tango, Arcus, Bioncotech, Compugen, CytomX, FLX Bio, ImaginAb, Isoplexis, Merus, Rgenix, and PACT.
SOURCE: Moyers J et al. AACR 2020, Abstract 4338.
Among patients with stage IIIC disease, the 2-year survival rate was 70% in those who received immunotherapy and 59% in those who did not (P < .01). The median overall survival in this group was 32.8 months with immunotherapy and 28 months without it (P < .01).
Among patients with stage IIIA disease, the 2-year survival rate was 94% with immunotherapy and 91% without it (P = .03).
There was a trend toward a 2-year survival benefit with immunotherapy in patients with stage IIIB disease and in all 4,094 stage III patients, but the differences were not significant. The 2-year survival rate was 84% with immunotherapy and 81% without it among patients with stage IIIB disease (P = .35). The survival rates were 83% and 80%, respectively, in all stage III patients (P = .051).
This was an early analysis, noted investigator Justin Moyers, MD, of Loma Linda (Calif.) University. Ipilimumab was approved as adjuvant therapy for stage III melanoma patients in 2015, the year patients from this analysis were diagnosed.
“There’s really only 2 full years of survival data,” Dr. Moyers said. “I think given time, we will see a benefit amongst all the substages.”
In the meantime, “I would definitely not use this data to say whether or not [immunotherapy] should be given,” Dr. Moyers said.
The researchers were just using the database – which captures 52% of U.S. melanoma cases – to see if “real-world data mimics the clinical trial data,” Dr. Moyers said.
Overall, the findings support “adjuvant immunotherapy in the real-world setting,” he said.
The researchers also looked at treatment patterns in 2015-2016 across 8,160 patients with stage III melanoma, 4,094 of whom were included in the aforementioned survival analysis. There were 2,260 patients (27.7%) who received immunotherapy after surgery during that time period.
Uptake of adjuvant immunotherapy “was low to start, but those patients did better than ones who did not get” it, said AACR president Antoni Ribas, MD, PhD, of the University of California, Los Angeles, who was not involved in the study.
Immunotherapy recipients were younger, on average (54.8 years vs. 62.4 years). Patients with Charlson comorbidity scores above zero and those on Medicare were less likely to receive immunotherapy (18.4% Medicare vs. over 30% with other payers). There also were trends of decreased use with lower income and lower high school graduation rates.
The finding “highlights the negative impact of socioeconomic [factors] on access to proven therapy,” Dr. Ribas said.
As for low use among Medicare patients, uptake of new treatments, in general, “seems to be faster with private insurance,” he noted.
The study excluded patients who received systemic therapies other than immunotherapy, as well as those who received immunotherapy before surgery. Among study limitations, the specific immunotherapies patients received was unknown.
There was no external funding for this study. Dr. Moyers reported travel compensation from Astellas Pharmaceuticals in 2018. Dr. Ribas disclosed relationships with Amgen, Chugai, Merck, Sanofi, Tango, Arcus, Bioncotech, Compugen, CytomX, FLX Bio, ImaginAb, Isoplexis, Merus, Rgenix, and PACT.
SOURCE: Moyers J et al. AACR 2020, Abstract 4338.
FROM AACR 2020
FDA approves new indications for pembrolizumab
The Food and Drug Administration recently announced two new types of cancer that can be treated by the anti–PD-1 antibody pembrolizumab.
The new indications expand the use of pembrolizumab (Keytruda) to include treatment of patients with unresectable or metastatic tumor mutational burden–high (TMB-H) solid tumors as well as patients with cutaneous squamous cell carcinoma (cSCC). The FDA announced the new indications just 8 days apart, on June 16 and June 24.
In addition, on June 29, the FDA approved a third new indication for pembrolizumab, this time as first-line treatment for patients with unresectable or metastatic microsatellite instability–high or mismatch repair–deficient colorectal cancer.
The new approvals add to a wide range of oncology indications for which pembrolizumab can be used.
Accelerated approval to treat solid tumors
The FDA granted accelerated approval for pembrolizumab to treat children and adults with unresectable or metastatic TMB-H solid tumors that progressed after previous treatment or in instances where there are no satisfactory alternative treatment options.
The tumor mutational burden must be confirmed by an FDA-approved test. To that end, the FDA approved the FoundationOneCDx assay, which is designed to help physicians determine which patients meet the threshold for TMB-H malignancies (10 or more mutations per megabase).
The efficacy of pembrolizumab in TMB-H solid tumors was investigated in 10 cohorts from the multicenter, open-label KEYNOTE-158 trial. Participants received 200 mg of pembrolizumab intravenously every 3 weeks until their disease progressed or they experienced unacceptable toxicity.
Within this population, 102 patients had tumors that met the TMB-H definition. In this group, the overall response rate was 29%, including a 25% partial response rate and a 4% complete response rate.
The median duration of response was not reached, but 57% of participants experienced a response lasting 12 months or longer, and 50% had a response lasting 24 months or longer.
The most common adverse events associated with pembrolizumab in this trial were fatigue, musculoskeletal pain, decreased appetite, pruritus, diarrhea, nausea, rash, pyrexia, cough, dyspnea, constipation, pain, and abdominal pain. Pembrolizumab is associated with immune-mediated side effects, including pneumonitis, colitis, hepatitis, endocrinopathies, nephritis, and skin adverse reactions, the FDA noted.
Safety and efficacy of pembrolizumab in pediatric patients with TMB-H central nervous system cancers have not been established.
New option for recurrent or metastatic cSCC
Physicians treating patients with cSCC that is not curable by surgery or radiation now have pembrolizumab to consider as another treatment option.
The cSCC approval is based on results of the multicenter, open-label KEYNOTE-629 trial. The dosage regimen was 200 mg of pembrolizumab intravenously every 3 weeks until cancer progressed, unacceptable toxicity arose, or 24 months of treatment were completed.
The objective response rate was 34%, and the median duration of response was not reached.
Adverse events were similar to those occurring in patients who received pembrolizumab as a single agent in other clinical trials, the FDA noted.
The Food and Drug Administration recently announced two new types of cancer that can be treated by the anti–PD-1 antibody pembrolizumab.
The new indications expand the use of pembrolizumab (Keytruda) to include treatment of patients with unresectable or metastatic tumor mutational burden–high (TMB-H) solid tumors as well as patients with cutaneous squamous cell carcinoma (cSCC). The FDA announced the new indications just 8 days apart, on June 16 and June 24.
In addition, on June 29, the FDA approved a third new indication for pembrolizumab, this time as first-line treatment for patients with unresectable or metastatic microsatellite instability–high or mismatch repair–deficient colorectal cancer.
The new approvals add to a wide range of oncology indications for which pembrolizumab can be used.
Accelerated approval to treat solid tumors
The FDA granted accelerated approval for pembrolizumab to treat children and adults with unresectable or metastatic TMB-H solid tumors that progressed after previous treatment or in instances where there are no satisfactory alternative treatment options.
The tumor mutational burden must be confirmed by an FDA-approved test. To that end, the FDA approved the FoundationOneCDx assay, which is designed to help physicians determine which patients meet the threshold for TMB-H malignancies (10 or more mutations per megabase).
The efficacy of pembrolizumab in TMB-H solid tumors was investigated in 10 cohorts from the multicenter, open-label KEYNOTE-158 trial. Participants received 200 mg of pembrolizumab intravenously every 3 weeks until their disease progressed or they experienced unacceptable toxicity.
Within this population, 102 patients had tumors that met the TMB-H definition. In this group, the overall response rate was 29%, including a 25% partial response rate and a 4% complete response rate.
The median duration of response was not reached, but 57% of participants experienced a response lasting 12 months or longer, and 50% had a response lasting 24 months or longer.
The most common adverse events associated with pembrolizumab in this trial were fatigue, musculoskeletal pain, decreased appetite, pruritus, diarrhea, nausea, rash, pyrexia, cough, dyspnea, constipation, pain, and abdominal pain. Pembrolizumab is associated with immune-mediated side effects, including pneumonitis, colitis, hepatitis, endocrinopathies, nephritis, and skin adverse reactions, the FDA noted.
Safety and efficacy of pembrolizumab in pediatric patients with TMB-H central nervous system cancers have not been established.
New option for recurrent or metastatic cSCC
Physicians treating patients with cSCC that is not curable by surgery or radiation now have pembrolizumab to consider as another treatment option.
The cSCC approval is based on results of the multicenter, open-label KEYNOTE-629 trial. The dosage regimen was 200 mg of pembrolizumab intravenously every 3 weeks until cancer progressed, unacceptable toxicity arose, or 24 months of treatment were completed.
The objective response rate was 34%, and the median duration of response was not reached.
Adverse events were similar to those occurring in patients who received pembrolizumab as a single agent in other clinical trials, the FDA noted.
The Food and Drug Administration recently announced two new types of cancer that can be treated by the anti–PD-1 antibody pembrolizumab.
The new indications expand the use of pembrolizumab (Keytruda) to include treatment of patients with unresectable or metastatic tumor mutational burden–high (TMB-H) solid tumors as well as patients with cutaneous squamous cell carcinoma (cSCC). The FDA announced the new indications just 8 days apart, on June 16 and June 24.
In addition, on June 29, the FDA approved a third new indication for pembrolizumab, this time as first-line treatment for patients with unresectable or metastatic microsatellite instability–high or mismatch repair–deficient colorectal cancer.
The new approvals add to a wide range of oncology indications for which pembrolizumab can be used.
Accelerated approval to treat solid tumors
The FDA granted accelerated approval for pembrolizumab to treat children and adults with unresectable or metastatic TMB-H solid tumors that progressed after previous treatment or in instances where there are no satisfactory alternative treatment options.
The tumor mutational burden must be confirmed by an FDA-approved test. To that end, the FDA approved the FoundationOneCDx assay, which is designed to help physicians determine which patients meet the threshold for TMB-H malignancies (10 or more mutations per megabase).
The efficacy of pembrolizumab in TMB-H solid tumors was investigated in 10 cohorts from the multicenter, open-label KEYNOTE-158 trial. Participants received 200 mg of pembrolizumab intravenously every 3 weeks until their disease progressed or they experienced unacceptable toxicity.
Within this population, 102 patients had tumors that met the TMB-H definition. In this group, the overall response rate was 29%, including a 25% partial response rate and a 4% complete response rate.
The median duration of response was not reached, but 57% of participants experienced a response lasting 12 months or longer, and 50% had a response lasting 24 months or longer.
The most common adverse events associated with pembrolizumab in this trial were fatigue, musculoskeletal pain, decreased appetite, pruritus, diarrhea, nausea, rash, pyrexia, cough, dyspnea, constipation, pain, and abdominal pain. Pembrolizumab is associated with immune-mediated side effects, including pneumonitis, colitis, hepatitis, endocrinopathies, nephritis, and skin adverse reactions, the FDA noted.
Safety and efficacy of pembrolizumab in pediatric patients with TMB-H central nervous system cancers have not been established.
New option for recurrent or metastatic cSCC
Physicians treating patients with cSCC that is not curable by surgery or radiation now have pembrolizumab to consider as another treatment option.
The cSCC approval is based on results of the multicenter, open-label KEYNOTE-629 trial. The dosage regimen was 200 mg of pembrolizumab intravenously every 3 weeks until cancer progressed, unacceptable toxicity arose, or 24 months of treatment were completed.
The objective response rate was 34%, and the median duration of response was not reached.
Adverse events were similar to those occurring in patients who received pembrolizumab as a single agent in other clinical trials, the FDA noted.
Personalized cancer vaccine may enhance checkpoint inhibitor activity
Combining a personalized cancer vaccine with an immune checkpoint inhibitor induced neoantigen-specific immune responses in most patients with advanced solid tumors in a phase 1b study.
Only two clinical responses were seen in this early investigation of the vaccine, RO7198457, combined with the PD-L1 inhibitor atezolizumab. However, T-cell responses were observed in about three-quarters of the patients evaluated, according to study investigator Juanita Lopez, MB BChir, PhD.
Those immune responses, coupled with preliminary evidence of infiltration of RO7198457-stimulated T cells into tumors, suggest the viability of this individualized anticancer strategy, according to Dr. Lopez, a consultant medical oncologist at The Royal Marsden NHS Foundation Trust and The Institute of Cancer Research, London.
“Failure of T-cell priming is a major cause of lack of response to immune checkpoint inhibitors,” Dr. Lopez said in an interview. “We hoped that, by eliciting a tumor-specific T-cell response, we would be able to overcome this.”
Preclinical data suggested the combination of vaccine and immune checkpoint inhibitors improved outcomes, which prompted the current study, added Dr. Lopez, who presented results from this study at the American Association for Cancer Research virtual meeting II.
Dr. Lopez noted that mutated neoantigens are recognized as foreign and have been shown to induce stronger T-cell responses, compared with shared antigens, likely because of a lack of central tolerance.
“Most of these mutated neoantigens are not shared between the patients, and therefore, targeted neoantigen-specific therapy requires an individualized approach,” she explained.
RO7198457 is manufactured on a per-patient basis and includes as many as 20 tumor-specific neoepitopes.
Study details
Dr. Lopez presented results from dose-escalation and expansion cohorts of the study, which included 142 patients with advanced solid tumors. The patients had colorectal, skin, kidney, lung, urothelial, breast, gynecologic, and head and neck cancers.
Most patients had low or no PD-L1 expression, and nearly 40% had received prior treatment with a checkpoint inhibitor.
Patients received nine doses of RO7198457 at 25-50 mcg during the 12-week induction stage. They then received RO7198457 every eight cycles until disease progression. Patients received atezolizumab at 1,200 mg on day 1 of each 21-day cycle.
Induction of proinflammatory cytokines was observed at each dose tested, and ex vivo T-cell responses were noted in 46 of 63 patients evaluated, or 73%.
T-cell receptors specific to RO7198457 were present posttreatment in a patient with rectal cancer, providing some preliminary evidence suggesting infiltration of RO7198457-stimulated T cells in the tumor, Dr. Lopez said.
There were two clinical responses. A patient with rectal cancer had a complete response, and a patient with triple-negative breast cancer had a partial response.
The combination of RO7198457 with atezolizumab was generally well tolerated, and the maximum tolerated dose was not reached, Dr. Lopez said. Most adverse events were grade 1/2, and immune-mediated adverse events were rare.
Implications and next steps
This study furthers earlier observations from neoantigen vaccine studies by linking dosing of the vaccine to dosing with immune checkpoint inhibitor, rather than giving the vaccine in the period leading up to immune checkpoint inhibitor administration, according to former AACR President Elaine R. Mardis, PhD, of Nationwide Children’s Hospital and The Ohio State University College of Medicine, both in Columbus.
That said, the implications for clinical practice remain unclear, according to Dr. Mardis.
“This combination did elicit an immune response that was highly specific for the neoantigen vaccine, but most patients did not receive a clinical benefit of disease response,” Dr. Mardis said in an interview. “This tells us the combination approach used was, overall, not quite right, and we need to continue to innovate in this area.”
The low clinical response rate in the study was likely caused in part by the fact that patients had very advanced disease and were heavily pretreated, according to Dr. Lopez
Randomized phase 2 studies of RO7198457 are now underway, Dr. Lopez said. One is a study of RO7198457 plus atezolizumab as adjuvant treatment for non–small cell lung cancer (NCT04267237). Another is testing RO7198457 in combination with pembrolizumab as first-line treatment for melanoma (NCT03815058).
The current study was funded by Genentech and BioNTech. Dr. Lopez reported disclosures related to Roche/Genentech, Basilea Pharmaceutica, and Genmab. Dr. Mardis reported disclosures related to Quiagen NV, PACT Pharma, Kiadis Pharma NV, and Interpreta.
SOURCE: Lopez J et al. AACR 2020, Abstract CT301.
Combining a personalized cancer vaccine with an immune checkpoint inhibitor induced neoantigen-specific immune responses in most patients with advanced solid tumors in a phase 1b study.
Only two clinical responses were seen in this early investigation of the vaccine, RO7198457, combined with the PD-L1 inhibitor atezolizumab. However, T-cell responses were observed in about three-quarters of the patients evaluated, according to study investigator Juanita Lopez, MB BChir, PhD.
Those immune responses, coupled with preliminary evidence of infiltration of RO7198457-stimulated T cells into tumors, suggest the viability of this individualized anticancer strategy, according to Dr. Lopez, a consultant medical oncologist at The Royal Marsden NHS Foundation Trust and The Institute of Cancer Research, London.
“Failure of T-cell priming is a major cause of lack of response to immune checkpoint inhibitors,” Dr. Lopez said in an interview. “We hoped that, by eliciting a tumor-specific T-cell response, we would be able to overcome this.”
Preclinical data suggested the combination of vaccine and immune checkpoint inhibitors improved outcomes, which prompted the current study, added Dr. Lopez, who presented results from this study at the American Association for Cancer Research virtual meeting II.
Dr. Lopez noted that mutated neoantigens are recognized as foreign and have been shown to induce stronger T-cell responses, compared with shared antigens, likely because of a lack of central tolerance.
“Most of these mutated neoantigens are not shared between the patients, and therefore, targeted neoantigen-specific therapy requires an individualized approach,” she explained.
RO7198457 is manufactured on a per-patient basis and includes as many as 20 tumor-specific neoepitopes.
Study details
Dr. Lopez presented results from dose-escalation and expansion cohorts of the study, which included 142 patients with advanced solid tumors. The patients had colorectal, skin, kidney, lung, urothelial, breast, gynecologic, and head and neck cancers.
Most patients had low or no PD-L1 expression, and nearly 40% had received prior treatment with a checkpoint inhibitor.
Patients received nine doses of RO7198457 at 25-50 mcg during the 12-week induction stage. They then received RO7198457 every eight cycles until disease progression. Patients received atezolizumab at 1,200 mg on day 1 of each 21-day cycle.
Induction of proinflammatory cytokines was observed at each dose tested, and ex vivo T-cell responses were noted in 46 of 63 patients evaluated, or 73%.
T-cell receptors specific to RO7198457 were present posttreatment in a patient with rectal cancer, providing some preliminary evidence suggesting infiltration of RO7198457-stimulated T cells in the tumor, Dr. Lopez said.
There were two clinical responses. A patient with rectal cancer had a complete response, and a patient with triple-negative breast cancer had a partial response.
The combination of RO7198457 with atezolizumab was generally well tolerated, and the maximum tolerated dose was not reached, Dr. Lopez said. Most adverse events were grade 1/2, and immune-mediated adverse events were rare.
Implications and next steps
This study furthers earlier observations from neoantigen vaccine studies by linking dosing of the vaccine to dosing with immune checkpoint inhibitor, rather than giving the vaccine in the period leading up to immune checkpoint inhibitor administration, according to former AACR President Elaine R. Mardis, PhD, of Nationwide Children’s Hospital and The Ohio State University College of Medicine, both in Columbus.
That said, the implications for clinical practice remain unclear, according to Dr. Mardis.
“This combination did elicit an immune response that was highly specific for the neoantigen vaccine, but most patients did not receive a clinical benefit of disease response,” Dr. Mardis said in an interview. “This tells us the combination approach used was, overall, not quite right, and we need to continue to innovate in this area.”
The low clinical response rate in the study was likely caused in part by the fact that patients had very advanced disease and were heavily pretreated, according to Dr. Lopez
Randomized phase 2 studies of RO7198457 are now underway, Dr. Lopez said. One is a study of RO7198457 plus atezolizumab as adjuvant treatment for non–small cell lung cancer (NCT04267237). Another is testing RO7198457 in combination with pembrolizumab as first-line treatment for melanoma (NCT03815058).
The current study was funded by Genentech and BioNTech. Dr. Lopez reported disclosures related to Roche/Genentech, Basilea Pharmaceutica, and Genmab. Dr. Mardis reported disclosures related to Quiagen NV, PACT Pharma, Kiadis Pharma NV, and Interpreta.
SOURCE: Lopez J et al. AACR 2020, Abstract CT301.
Combining a personalized cancer vaccine with an immune checkpoint inhibitor induced neoantigen-specific immune responses in most patients with advanced solid tumors in a phase 1b study.
Only two clinical responses were seen in this early investigation of the vaccine, RO7198457, combined with the PD-L1 inhibitor atezolizumab. However, T-cell responses were observed in about three-quarters of the patients evaluated, according to study investigator Juanita Lopez, MB BChir, PhD.
Those immune responses, coupled with preliminary evidence of infiltration of RO7198457-stimulated T cells into tumors, suggest the viability of this individualized anticancer strategy, according to Dr. Lopez, a consultant medical oncologist at The Royal Marsden NHS Foundation Trust and The Institute of Cancer Research, London.
“Failure of T-cell priming is a major cause of lack of response to immune checkpoint inhibitors,” Dr. Lopez said in an interview. “We hoped that, by eliciting a tumor-specific T-cell response, we would be able to overcome this.”
Preclinical data suggested the combination of vaccine and immune checkpoint inhibitors improved outcomes, which prompted the current study, added Dr. Lopez, who presented results from this study at the American Association for Cancer Research virtual meeting II.
Dr. Lopez noted that mutated neoantigens are recognized as foreign and have been shown to induce stronger T-cell responses, compared with shared antigens, likely because of a lack of central tolerance.
“Most of these mutated neoantigens are not shared between the patients, and therefore, targeted neoantigen-specific therapy requires an individualized approach,” she explained.
RO7198457 is manufactured on a per-patient basis and includes as many as 20 tumor-specific neoepitopes.
Study details
Dr. Lopez presented results from dose-escalation and expansion cohorts of the study, which included 142 patients with advanced solid tumors. The patients had colorectal, skin, kidney, lung, urothelial, breast, gynecologic, and head and neck cancers.
Most patients had low or no PD-L1 expression, and nearly 40% had received prior treatment with a checkpoint inhibitor.
Patients received nine doses of RO7198457 at 25-50 mcg during the 12-week induction stage. They then received RO7198457 every eight cycles until disease progression. Patients received atezolizumab at 1,200 mg on day 1 of each 21-day cycle.
Induction of proinflammatory cytokines was observed at each dose tested, and ex vivo T-cell responses were noted in 46 of 63 patients evaluated, or 73%.
T-cell receptors specific to RO7198457 were present posttreatment in a patient with rectal cancer, providing some preliminary evidence suggesting infiltration of RO7198457-stimulated T cells in the tumor, Dr. Lopez said.
There were two clinical responses. A patient with rectal cancer had a complete response, and a patient with triple-negative breast cancer had a partial response.
The combination of RO7198457 with atezolizumab was generally well tolerated, and the maximum tolerated dose was not reached, Dr. Lopez said. Most adverse events were grade 1/2, and immune-mediated adverse events were rare.
Implications and next steps
This study furthers earlier observations from neoantigen vaccine studies by linking dosing of the vaccine to dosing with immune checkpoint inhibitor, rather than giving the vaccine in the period leading up to immune checkpoint inhibitor administration, according to former AACR President Elaine R. Mardis, PhD, of Nationwide Children’s Hospital and The Ohio State University College of Medicine, both in Columbus.
That said, the implications for clinical practice remain unclear, according to Dr. Mardis.
“This combination did elicit an immune response that was highly specific for the neoantigen vaccine, but most patients did not receive a clinical benefit of disease response,” Dr. Mardis said in an interview. “This tells us the combination approach used was, overall, not quite right, and we need to continue to innovate in this area.”
The low clinical response rate in the study was likely caused in part by the fact that patients had very advanced disease and were heavily pretreated, according to Dr. Lopez
Randomized phase 2 studies of RO7198457 are now underway, Dr. Lopez said. One is a study of RO7198457 plus atezolizumab as adjuvant treatment for non–small cell lung cancer (NCT04267237). Another is testing RO7198457 in combination with pembrolizumab as first-line treatment for melanoma (NCT03815058).
The current study was funded by Genentech and BioNTech. Dr. Lopez reported disclosures related to Roche/Genentech, Basilea Pharmaceutica, and Genmab. Dr. Mardis reported disclosures related to Quiagen NV, PACT Pharma, Kiadis Pharma NV, and Interpreta.
SOURCE: Lopez J et al. AACR 2020, Abstract CT301.
FROM AACR 2020
Study evaluates number of needed to refer, biopsy for diagnosing a melanoma
At the same time, the number needed to refer to diagnose non-melanoma skin cancer was 4 and the number needed to biopsy was 1.5.
The findings come from a retrospective review of 707 patients referred to a tertiary medical center dermatology practice for suspicious lesions, presented in a poster session at the virtual annual meeting of the American Academy of Dermatology
“Multiple studies in the dermatology literature have looked at the number needed to treat (NNT) as a quality metric for dermatology clinics, where a lower number is ‘better,’” the study’s first author, Nikolai Klebanov, MD, said in an interview following the virtual meeting. “Our particular study is unique in that we estimated both the number needed to refer and number needed to biopsy to closely examine the process of referrals for suspicious lesions from primary care settings to specialists. We also looked closely at the underlying patient-centered characteristics, which could be used by all clinicians to streamline the referral process by reducing the volume of low-risk referrals.”
Dr. Klebanov, of the department of dermatology at Massachusetts General Hospital, Boston, and his associates reviewed 707 unique patient visits to the department during July 2015–February 2016. They calculated the number needed to refer and biopsy for melanoma as the ratio of biopsy-proven melanoma diagnoses among benign and dysplastic nevi and seborrheic keratoses. For nonmelanoma skin cancer, they used the ratio of basal and squamous cell carcinoma among actinic keratoses and seborrheic keratoses.
Of the 707 patients, 54% were female, and males were slightly older than females (a mean of 58 vs. 54 years, respectively). The researchers found that lesions were more commonly benign among all age groups, while the frequency of premalignant and malignant lesions such as actinic keratoses, nonmelanoma skin cancer, and melanoma was highest for males and increased with age. Nevi were the most common benign diagnosis among patients 39 years of age and younger, while seborrheic keratoses were more common among patients aged 40 years and older.
The researchers found that the number needed to treat for melanoma was 31.5 and the number needed to biopsy was 7.5, which represents a 4.2-fold difference. Meanwhile, the number needed to refer for nonmelanoma skin cancer was 4, and the number needed to biopsy was 1.5, which represents a 2.7-fold difference. Despite variable rates of skin cancer between demographics, the biopsy rate ranged between 18% and 30%, for a mean of 23.4%.
“We found that most young patients referred for a ‘suspicious lesion’ on clinical prebiopsy assessment by the dermatologist were determined to actually have a benign nevus, and that older patients were most likely to have a seborrheic keratosis as the underlying lesion,” Dr. Klebanov said. “Among the minority of patients in each demographic group who were selected for biopsy, those lesions which were found to be benign were also largely nevi and keratoses. Even by being mindful of just the patient’s age, primary care providers can follow patients clinically with a tailored differential diagnosis in mind before referral, and dermatologists can reduce the number of biopsies they perform on patients who are being referred.”
He added that he and his colleagues were surprised that despite very low rates of skin cancer in young patients, and thus different pretest probabilities of cancer, biopsy rates across demographics were consistently around 20%. “We also found a disproportionate number of female patients younger than age 40 who were referred for suspicious lesions, while in the older age groups, the ratio of males to females was approximately equal.”
Dr. Klebanov acknowledged certain limitations of the study, including its single-center, retrospective design, and that information was not collected on patients’ family history of skin cancer, Fitzpatrick skin type, nor the clinical course of the lesion while it was followed by the primary care office. “The nuanced differences in these factors may certainly play a role in decisions for individual patients,” he said.
The study’s principal investigator was Hensin Tsao MD, PhD, clinical director of the MGH Melanoma & Pigmented Lesion Center The work was supported by the Alpha Omega Alpha Carolyn Kuckein Research Fellowship. The researchers reported having no financial disclosures.
SOURCE: Klebanov N et al. AAD 20. Abstract 15881.
At the same time, the number needed to refer to diagnose non-melanoma skin cancer was 4 and the number needed to biopsy was 1.5.
The findings come from a retrospective review of 707 patients referred to a tertiary medical center dermatology practice for suspicious lesions, presented in a poster session at the virtual annual meeting of the American Academy of Dermatology
“Multiple studies in the dermatology literature have looked at the number needed to treat (NNT) as a quality metric for dermatology clinics, where a lower number is ‘better,’” the study’s first author, Nikolai Klebanov, MD, said in an interview following the virtual meeting. “Our particular study is unique in that we estimated both the number needed to refer and number needed to biopsy to closely examine the process of referrals for suspicious lesions from primary care settings to specialists. We also looked closely at the underlying patient-centered characteristics, which could be used by all clinicians to streamline the referral process by reducing the volume of low-risk referrals.”
Dr. Klebanov, of the department of dermatology at Massachusetts General Hospital, Boston, and his associates reviewed 707 unique patient visits to the department during July 2015–February 2016. They calculated the number needed to refer and biopsy for melanoma as the ratio of biopsy-proven melanoma diagnoses among benign and dysplastic nevi and seborrheic keratoses. For nonmelanoma skin cancer, they used the ratio of basal and squamous cell carcinoma among actinic keratoses and seborrheic keratoses.
Of the 707 patients, 54% were female, and males were slightly older than females (a mean of 58 vs. 54 years, respectively). The researchers found that lesions were more commonly benign among all age groups, while the frequency of premalignant and malignant lesions such as actinic keratoses, nonmelanoma skin cancer, and melanoma was highest for males and increased with age. Nevi were the most common benign diagnosis among patients 39 years of age and younger, while seborrheic keratoses were more common among patients aged 40 years and older.
The researchers found that the number needed to treat for melanoma was 31.5 and the number needed to biopsy was 7.5, which represents a 4.2-fold difference. Meanwhile, the number needed to refer for nonmelanoma skin cancer was 4, and the number needed to biopsy was 1.5, which represents a 2.7-fold difference. Despite variable rates of skin cancer between demographics, the biopsy rate ranged between 18% and 30%, for a mean of 23.4%.
“We found that most young patients referred for a ‘suspicious lesion’ on clinical prebiopsy assessment by the dermatologist were determined to actually have a benign nevus, and that older patients were most likely to have a seborrheic keratosis as the underlying lesion,” Dr. Klebanov said. “Among the minority of patients in each demographic group who were selected for biopsy, those lesions which were found to be benign were also largely nevi and keratoses. Even by being mindful of just the patient’s age, primary care providers can follow patients clinically with a tailored differential diagnosis in mind before referral, and dermatologists can reduce the number of biopsies they perform on patients who are being referred.”
He added that he and his colleagues were surprised that despite very low rates of skin cancer in young patients, and thus different pretest probabilities of cancer, biopsy rates across demographics were consistently around 20%. “We also found a disproportionate number of female patients younger than age 40 who were referred for suspicious lesions, while in the older age groups, the ratio of males to females was approximately equal.”
Dr. Klebanov acknowledged certain limitations of the study, including its single-center, retrospective design, and that information was not collected on patients’ family history of skin cancer, Fitzpatrick skin type, nor the clinical course of the lesion while it was followed by the primary care office. “The nuanced differences in these factors may certainly play a role in decisions for individual patients,” he said.
The study’s principal investigator was Hensin Tsao MD, PhD, clinical director of the MGH Melanoma & Pigmented Lesion Center The work was supported by the Alpha Omega Alpha Carolyn Kuckein Research Fellowship. The researchers reported having no financial disclosures.
SOURCE: Klebanov N et al. AAD 20. Abstract 15881.
At the same time, the number needed to refer to diagnose non-melanoma skin cancer was 4 and the number needed to biopsy was 1.5.
The findings come from a retrospective review of 707 patients referred to a tertiary medical center dermatology practice for suspicious lesions, presented in a poster session at the virtual annual meeting of the American Academy of Dermatology
“Multiple studies in the dermatology literature have looked at the number needed to treat (NNT) as a quality metric for dermatology clinics, where a lower number is ‘better,’” the study’s first author, Nikolai Klebanov, MD, said in an interview following the virtual meeting. “Our particular study is unique in that we estimated both the number needed to refer and number needed to biopsy to closely examine the process of referrals for suspicious lesions from primary care settings to specialists. We also looked closely at the underlying patient-centered characteristics, which could be used by all clinicians to streamline the referral process by reducing the volume of low-risk referrals.”
Dr. Klebanov, of the department of dermatology at Massachusetts General Hospital, Boston, and his associates reviewed 707 unique patient visits to the department during July 2015–February 2016. They calculated the number needed to refer and biopsy for melanoma as the ratio of biopsy-proven melanoma diagnoses among benign and dysplastic nevi and seborrheic keratoses. For nonmelanoma skin cancer, they used the ratio of basal and squamous cell carcinoma among actinic keratoses and seborrheic keratoses.
Of the 707 patients, 54% were female, and males were slightly older than females (a mean of 58 vs. 54 years, respectively). The researchers found that lesions were more commonly benign among all age groups, while the frequency of premalignant and malignant lesions such as actinic keratoses, nonmelanoma skin cancer, and melanoma was highest for males and increased with age. Nevi were the most common benign diagnosis among patients 39 years of age and younger, while seborrheic keratoses were more common among patients aged 40 years and older.
The researchers found that the number needed to treat for melanoma was 31.5 and the number needed to biopsy was 7.5, which represents a 4.2-fold difference. Meanwhile, the number needed to refer for nonmelanoma skin cancer was 4, and the number needed to biopsy was 1.5, which represents a 2.7-fold difference. Despite variable rates of skin cancer between demographics, the biopsy rate ranged between 18% and 30%, for a mean of 23.4%.
“We found that most young patients referred for a ‘suspicious lesion’ on clinical prebiopsy assessment by the dermatologist were determined to actually have a benign nevus, and that older patients were most likely to have a seborrheic keratosis as the underlying lesion,” Dr. Klebanov said. “Among the minority of patients in each demographic group who were selected for biopsy, those lesions which were found to be benign were also largely nevi and keratoses. Even by being mindful of just the patient’s age, primary care providers can follow patients clinically with a tailored differential diagnosis in mind before referral, and dermatologists can reduce the number of biopsies they perform on patients who are being referred.”
He added that he and his colleagues were surprised that despite very low rates of skin cancer in young patients, and thus different pretest probabilities of cancer, biopsy rates across demographics were consistently around 20%. “We also found a disproportionate number of female patients younger than age 40 who were referred for suspicious lesions, while in the older age groups, the ratio of males to females was approximately equal.”
Dr. Klebanov acknowledged certain limitations of the study, including its single-center, retrospective design, and that information was not collected on patients’ family history of skin cancer, Fitzpatrick skin type, nor the clinical course of the lesion while it was followed by the primary care office. “The nuanced differences in these factors may certainly play a role in decisions for individual patients,” he said.
The study’s principal investigator was Hensin Tsao MD, PhD, clinical director of the MGH Melanoma & Pigmented Lesion Center The work was supported by the Alpha Omega Alpha Carolyn Kuckein Research Fellowship. The researchers reported having no financial disclosures.
SOURCE: Klebanov N et al. AAD 20. Abstract 15881.
FROM AAD 20
Vulvar melanoma is increasing in older women
Maia K. Erickson reported in a poster at the virtual annual meeting of the American Academy of Dermatology.
These are often aggressive malignancies. The 5-year survival following diagnosis of vulvar melanoma in women aged 60 years or older was 39.7%, compared with 61.9% in younger women, according to Ms. Erickson, a visiting research fellow in the department of dermatology at Northwestern University, Chicago.
She presented a population-based study of epidemiologic trends in vulvar melanoma based upon analysis of the National Cancer Institute’s Surveillance, Epidemiology and End Results database. Vulvar melanoma was rare during the study years 2000-2016, with an overall incidence rate of 0.1 cases per 100,000 women. That worked out to 746 analyzable cases. Of note, the incidence rate ratio was 680% higher in older women (age 60 and older).
One reason for the markedly worse 5-year survival in older women was that the predominant histologic subtype of vulvar melanoma in that population was nodular melanoma, accounting for 48% of the cases where a histologic subtype was specified. In contrast, the less-aggressive superficial spreading melanoma subtype prevailed in patients aged under 60 years, accounting for 63% of cases.
About 93% of vulvar melanomas occurred in whites; 63% were local and 8.7% were metastatic.
Ms. Erickson noted that the vulva is the most common site for gynecologic tract melanomas, accounting for 70% of them. And while the female genitalia make up only 1%-2% of body surface area, that’s the anatomic site of up to 7% of all melanomas in women.
She reported having no financial conflicts regarding her study.
Maia K. Erickson reported in a poster at the virtual annual meeting of the American Academy of Dermatology.
These are often aggressive malignancies. The 5-year survival following diagnosis of vulvar melanoma in women aged 60 years or older was 39.7%, compared with 61.9% in younger women, according to Ms. Erickson, a visiting research fellow in the department of dermatology at Northwestern University, Chicago.
She presented a population-based study of epidemiologic trends in vulvar melanoma based upon analysis of the National Cancer Institute’s Surveillance, Epidemiology and End Results database. Vulvar melanoma was rare during the study years 2000-2016, with an overall incidence rate of 0.1 cases per 100,000 women. That worked out to 746 analyzable cases. Of note, the incidence rate ratio was 680% higher in older women (age 60 and older).
One reason for the markedly worse 5-year survival in older women was that the predominant histologic subtype of vulvar melanoma in that population was nodular melanoma, accounting for 48% of the cases where a histologic subtype was specified. In contrast, the less-aggressive superficial spreading melanoma subtype prevailed in patients aged under 60 years, accounting for 63% of cases.
About 93% of vulvar melanomas occurred in whites; 63% were local and 8.7% were metastatic.
Ms. Erickson noted that the vulva is the most common site for gynecologic tract melanomas, accounting for 70% of them. And while the female genitalia make up only 1%-2% of body surface area, that’s the anatomic site of up to 7% of all melanomas in women.
She reported having no financial conflicts regarding her study.
Maia K. Erickson reported in a poster at the virtual annual meeting of the American Academy of Dermatology.
These are often aggressive malignancies. The 5-year survival following diagnosis of vulvar melanoma in women aged 60 years or older was 39.7%, compared with 61.9% in younger women, according to Ms. Erickson, a visiting research fellow in the department of dermatology at Northwestern University, Chicago.
She presented a population-based study of epidemiologic trends in vulvar melanoma based upon analysis of the National Cancer Institute’s Surveillance, Epidemiology and End Results database. Vulvar melanoma was rare during the study years 2000-2016, with an overall incidence rate of 0.1 cases per 100,000 women. That worked out to 746 analyzable cases. Of note, the incidence rate ratio was 680% higher in older women (age 60 and older).
One reason for the markedly worse 5-year survival in older women was that the predominant histologic subtype of vulvar melanoma in that population was nodular melanoma, accounting for 48% of the cases where a histologic subtype was specified. In contrast, the less-aggressive superficial spreading melanoma subtype prevailed in patients aged under 60 years, accounting for 63% of cases.
About 93% of vulvar melanomas occurred in whites; 63% were local and 8.7% were metastatic.
Ms. Erickson noted that the vulva is the most common site for gynecologic tract melanomas, accounting for 70% of them. And while the female genitalia make up only 1%-2% of body surface area, that’s the anatomic site of up to 7% of all melanomas in women.
She reported having no financial conflicts regarding her study.
FROM AAD 2020
Combo exhibits activity in metastatic mucosal melanoma
a presentation made as part of the American Society of Clinical Oncology virtual scientific program.
according toThe combination was well tolerated and “the preliminary efficacy seems to be promising,” which warrants a phase 3 trial, said investigator Jun Guo, MD, of the Peking University Cancer Hospital and Institute in Beijing, who presented the findings.
Mucosal melanoma does not respond as well as cutaneous melanoma to standard programmed death-1 (PD-1) blockade, so investigators are looking for additional options, Dr. Guo noted. Earlier studies have shown that vascular endothelial growth factor expression correlates negatively with clinical outcome, so the combination of VEGF inhibition with PD-1 blockade might provide therapeutic opportunities.
To find out, Dr. Guo and colleagues tested the anti-PD-1 antibody toripalimab in combination with the VEGF inhibitor axitinib in a phase 1 trial. The trial was conducted in China, where mucosal melanoma accounts for up to a quarter of all melanoma cases and where toripalimab is approved to treat mucosal melanoma.
The trial enrolled 33 patients with pathologically confirmed metastatic mucosal melanoma. The esophagus and genital tract were the most common primary lesion sites (both 21.2%). The patients’ average age was 53.4 years, and 60.6% were women. Two patients (6.1%) had previously received systemic chemotherapy. Most (64.6%) were PD–ligand 1 (PD-L1) negative, and most (60.6%) were BRAF/RAS/NF1 wild type.
The patients received axitinib at 5 mg twice daily plus toripalimab at 3 mg/kg every 2 weeks until confirmed disease progression, unacceptable toxicity, or voluntary withdrawal.
As of May 2, 2020, the overall response rate was 48.5%. There were 15 partial responses and 1 complete response. The median duration of response was 13.7 months. The median progression-free survival was 7.5 months, and the median overall survival was 20.7 months.
Progression-free and overall survival were numerically higher in PD-L1-positive subjects and those with higher tumor mutation burdens. An expression profile of 12 genes related to inflammation and angiogenesis showed a significant correlation with response. This might help identify patients most likely to respond to the combination, but further validation is needed, Dr. Guo said.
A total of 32 subjects (97%) have had a treatment-related adverse event, including 13 (39.4%) with grade 3-5 events. The most common of these were proteinuria, hypertension, and neutropenia (all 9.1%).
“So does this study address the unmet need? In many ways, yes,” said Ryan Sullivan, MD, an assistant professor of hematology/oncology at Massachusetts General Hospital in Boston, and the discussant on Dr. Guo’s presentation.
“However, the data to date [don’t] mean we should be treating all of our mucosal melanoma patients with axitinib plus an anti-PD-1 antibody. There needs to be randomized data, but I would describe this data as very encouraging,” he said.
The study was funded by the maker of toripalimab, Shanghai Junshi Bioscience. Dr. Guo disclosed relationships with Shanghai Junshi Bioscience and Pfizer, maker of axitinib. Other investigators are employed by Shanghai Junshi Bioscience. Dr. Sullivan reported institutional research funding from Pfizer.
SOURCE: Guo J et al. ASCO 2020, Abstract 10007.
a presentation made as part of the American Society of Clinical Oncology virtual scientific program.
according toThe combination was well tolerated and “the preliminary efficacy seems to be promising,” which warrants a phase 3 trial, said investigator Jun Guo, MD, of the Peking University Cancer Hospital and Institute in Beijing, who presented the findings.
Mucosal melanoma does not respond as well as cutaneous melanoma to standard programmed death-1 (PD-1) blockade, so investigators are looking for additional options, Dr. Guo noted. Earlier studies have shown that vascular endothelial growth factor expression correlates negatively with clinical outcome, so the combination of VEGF inhibition with PD-1 blockade might provide therapeutic opportunities.
To find out, Dr. Guo and colleagues tested the anti-PD-1 antibody toripalimab in combination with the VEGF inhibitor axitinib in a phase 1 trial. The trial was conducted in China, where mucosal melanoma accounts for up to a quarter of all melanoma cases and where toripalimab is approved to treat mucosal melanoma.
The trial enrolled 33 patients with pathologically confirmed metastatic mucosal melanoma. The esophagus and genital tract were the most common primary lesion sites (both 21.2%). The patients’ average age was 53.4 years, and 60.6% were women. Two patients (6.1%) had previously received systemic chemotherapy. Most (64.6%) were PD–ligand 1 (PD-L1) negative, and most (60.6%) were BRAF/RAS/NF1 wild type.
The patients received axitinib at 5 mg twice daily plus toripalimab at 3 mg/kg every 2 weeks until confirmed disease progression, unacceptable toxicity, or voluntary withdrawal.
As of May 2, 2020, the overall response rate was 48.5%. There were 15 partial responses and 1 complete response. The median duration of response was 13.7 months. The median progression-free survival was 7.5 months, and the median overall survival was 20.7 months.
Progression-free and overall survival were numerically higher in PD-L1-positive subjects and those with higher tumor mutation burdens. An expression profile of 12 genes related to inflammation and angiogenesis showed a significant correlation with response. This might help identify patients most likely to respond to the combination, but further validation is needed, Dr. Guo said.
A total of 32 subjects (97%) have had a treatment-related adverse event, including 13 (39.4%) with grade 3-5 events. The most common of these were proteinuria, hypertension, and neutropenia (all 9.1%).
“So does this study address the unmet need? In many ways, yes,” said Ryan Sullivan, MD, an assistant professor of hematology/oncology at Massachusetts General Hospital in Boston, and the discussant on Dr. Guo’s presentation.
“However, the data to date [don’t] mean we should be treating all of our mucosal melanoma patients with axitinib plus an anti-PD-1 antibody. There needs to be randomized data, but I would describe this data as very encouraging,” he said.
The study was funded by the maker of toripalimab, Shanghai Junshi Bioscience. Dr. Guo disclosed relationships with Shanghai Junshi Bioscience and Pfizer, maker of axitinib. Other investigators are employed by Shanghai Junshi Bioscience. Dr. Sullivan reported institutional research funding from Pfizer.
SOURCE: Guo J et al. ASCO 2020, Abstract 10007.
a presentation made as part of the American Society of Clinical Oncology virtual scientific program.
according toThe combination was well tolerated and “the preliminary efficacy seems to be promising,” which warrants a phase 3 trial, said investigator Jun Guo, MD, of the Peking University Cancer Hospital and Institute in Beijing, who presented the findings.
Mucosal melanoma does not respond as well as cutaneous melanoma to standard programmed death-1 (PD-1) blockade, so investigators are looking for additional options, Dr. Guo noted. Earlier studies have shown that vascular endothelial growth factor expression correlates negatively with clinical outcome, so the combination of VEGF inhibition with PD-1 blockade might provide therapeutic opportunities.
To find out, Dr. Guo and colleagues tested the anti-PD-1 antibody toripalimab in combination with the VEGF inhibitor axitinib in a phase 1 trial. The trial was conducted in China, where mucosal melanoma accounts for up to a quarter of all melanoma cases and where toripalimab is approved to treat mucosal melanoma.
The trial enrolled 33 patients with pathologically confirmed metastatic mucosal melanoma. The esophagus and genital tract were the most common primary lesion sites (both 21.2%). The patients’ average age was 53.4 years, and 60.6% were women. Two patients (6.1%) had previously received systemic chemotherapy. Most (64.6%) were PD–ligand 1 (PD-L1) negative, and most (60.6%) were BRAF/RAS/NF1 wild type.
The patients received axitinib at 5 mg twice daily plus toripalimab at 3 mg/kg every 2 weeks until confirmed disease progression, unacceptable toxicity, or voluntary withdrawal.
As of May 2, 2020, the overall response rate was 48.5%. There were 15 partial responses and 1 complete response. The median duration of response was 13.7 months. The median progression-free survival was 7.5 months, and the median overall survival was 20.7 months.
Progression-free and overall survival were numerically higher in PD-L1-positive subjects and those with higher tumor mutation burdens. An expression profile of 12 genes related to inflammation and angiogenesis showed a significant correlation with response. This might help identify patients most likely to respond to the combination, but further validation is needed, Dr. Guo said.
A total of 32 subjects (97%) have had a treatment-related adverse event, including 13 (39.4%) with grade 3-5 events. The most common of these were proteinuria, hypertension, and neutropenia (all 9.1%).
“So does this study address the unmet need? In many ways, yes,” said Ryan Sullivan, MD, an assistant professor of hematology/oncology at Massachusetts General Hospital in Boston, and the discussant on Dr. Guo’s presentation.
“However, the data to date [don’t] mean we should be treating all of our mucosal melanoma patients with axitinib plus an anti-PD-1 antibody. There needs to be randomized data, but I would describe this data as very encouraging,” he said.
The study was funded by the maker of toripalimab, Shanghai Junshi Bioscience. Dr. Guo disclosed relationships with Shanghai Junshi Bioscience and Pfizer, maker of axitinib. Other investigators are employed by Shanghai Junshi Bioscience. Dr. Sullivan reported institutional research funding from Pfizer.
SOURCE: Guo J et al. ASCO 2020, Abstract 10007.
FROM ASCO 2020
Anti–PD1 Immune Checkpoint Inhibitor–Induced Bullous Pemphigoid in Metastatic Melanoma and Non–Small Cell Lung Cancer
Immune checkpoint inhibitors are used for a variety of advanced malignancies, including melanoma, non–small cell lung cancer, urothelial cancer, and renal cell carcinoma. Anti–programmed cell death 1 (PD1) targeted therapies, such as pembrolizumab and nivolumab, are improving patient survival. This class of immunotherapy is revolutionary but is associated with autoimmune adverse effects. A rare but increasingly reported adverse effect of anti-PD1 therapy is bullous pemphigoid (BP), an autoimmune blistering disease directed against
High clinical suspicion, early diagnosis, and proper management of immunotherapy-related BP are imperative for keeping patients on life-prolonging treatment. We present 3 cases of BP secondary to anti-PD1 immunotherapy in patients with melanoma or non–small cell lung cancer to highlight the diagnosis and treatment of BP as well as emphasize the importance of the dermatologist in the care of patients with immunotherapy-related skin disease.
Case Reports
Patient 1
A 72-year-old woman with metastatic BRAF-mutated melanoma from an unknown primary site presented with intensely pruritic papules on the back, chest, and extremities of 4 months’ duration. She described her symptoms as insidious in onset and refractory to clobetasol ointment, oral diphenhydramine, and over-the-counter anti-itch creams. The patient had been treated with oral dabrafenib 150 mg twice daily and trametinib 2 mg/d but was switched to pembrolizumab when the disease progressed. After 8 months, she had a complete radiologic response to pembrolizumab 2 mg/kg every 3 weeks, which was discontinued in favor of observation 3 months prior to presentation to dermatology.
At the current presentation, physical examination revealed innumerable erythematous, excoriated, 2- to 4-mm, red papules diffusely scattered on the upper back, chest, abdomen, and thighs, with one 8×4-mm vesicle on the right side of the upper back (Figure 1). Discrete areas of depigmented macules, consistent with vitiligo, coalesced into patches on the legs, thighs, arms, and back. The patient was started on a 3-week oral prednisone taper for symptom relief. A hematoxylin and eosin (H&E)–stained punch biopsy of the back revealed a subepidermal split with eosinophils and a dense eosinophilic infiltrate in the dermis (Figure 2). Direct immunofluorescence (DIF) studies from a specimen adjacent to the biopsy collected for H&E staining showed linear deposition of IgA, IgG, and C3 along the dermoepidermal junction (Figure 3). Histologic findings were consistent with BP.
The patient was started on doxycycline 100 mg twice daily and clobetasol ointment 0.05% once daily to supplement the prednisone taper. At 3-week follow-up, she reported pruritus and a few erythematous macules but no new bullae. At 12 weeks, some papules persisted; however, the patient was averse to using systemic agents and decided that symptoms were adequately controlled with clobetasol ointment and oral doxycycline.
Because the patient currently remains in clinical and radiologic remission, anti-PD1 immune checkpoint inhibitors have not been restarted but remain an option for the future if disease recurs
Patient 2
An 82-year-old man with a history of stage IIC desmoplastic melanoma presented to dermatology with an intensely pruritic eruption on the legs, arms, waist, upper torso, and scalp of 3 weeks’ duration. Clobetasol ointment had provided minimal relief.
Six months prior to presenting to dermatology, the patient underwent immunotherapy with 4 cycles of ipilimumab 200 mg intravenous (IV) and nivolumab 240 mg IV every 2 weeks, receiving ipilimumab during the first cycle only because of a lack of availability at the pharmacy. He then received nivolumab 240 mg IV every 2 weeks as maintenance therapy. After the second dose of nivolumab maintenance therapy, however, he developed generalized bullae and pruritus. Dermatology was consulted during an oncology appointment, and his oncologist decided to hold nivolumab.
Physical examination revealed generalized tense and eroded bullae covering more than 50% of the body surface area and affecting the scalp, arms, legs, torso, and buttocks. Two punch biopsies were obtained. Hematoxylin and eosin staining revealed a subepidermal split with predominantly eosinophils and scattered neutrophils. Direct immunofluorescence studies showed linear deposition of IgG, IgA, and C3 along the dermoepidermal junction, consistent with BP.
The patient’s BP was difficult to control, requiring several hospital admissions for wound care, high-dose systemic steroids, and initiation of mycophenolate mofetil. After 4 months of waxing and waning symptoms, the BP was controlled with mycophenolate mofetil 1500 mg/d; clobetasol ointment 0.05%; and diphenhydramine for pruritus. Due to the prolonged recovery and severity of BP, the patient’s oncologist deemed that he was not a candidate for future immunotherapy.
Patient 3
A 68-year-old man with PD1-negative, metastatic, well-differentiated squamous cell carcinoma of the lung presented to dermatology with a pruritic rash of 3 weeks’ duration. He had been receiving nivolumab for 2 years after disease progressed on prior chemotherapies and experienced several grade 1 or grade 2 nivolumab-induced autoimmune reactions including thyroiditis, dermatitis, and nephritis, for which he was taking prednisone 5 mg/d for suppression.
Physical examination revealed psoriasiform pink plaques on the arms, chest, and legs. The differential diagnosis at the time favored psoriasiform dermatitis over lichenoid dermatitis. A punch biopsy revealed psoriasiform dermatitis. The patient was prescribed fluocinonide ointment 0.05% daily. His plaques improved with topical steroids.
The patient returned approximately 1 month later with a report of a new blistering rash on the legs. Physical examination revealed interval improvement of the psoriasiform plaques on the scalp, torso, and extremities, but tense bullae were seen on the thighs, with surrounding superficial erosions at sites of recent bullae. Punch biopsies of the skin for H&E staining and DIF showed BP.
Prednisone was increased to 50 mg/d for a 3-week taper. Doxycycline 100 mg twice daily was started. The patient’s skin disease continued to be difficult to control with therapy; nivolumab was held by his oncologist.
Comment
Immunotherapy with immune checkpoint blockade represents a successful application of immune recognition to treat metastatic cancers, including melanoma, non–small cell lung cancer, urothelial cancer, and renal cell carcinoma.
Anti-PD1 targeted therapies improve survival in solid and hematologic malignancies, with a response rate as high as 40% in melanoma.2 Although these medications can prolong survival, many are associated with loss of self-tolerance and severe autoimmunelike events that can limit therapy.3 An exception is PD1-induced vitiligo, which patient 1 developed and has been associated with a better response to therapy.4
Anti-PD1–induced BP is a newly reported adverse effect. In its early stages, BP can be difficult to differentiate from eczematous or urticarial dermatitis.5-8 Discontinuation of immunotherapy has been reported in more than 70% of patients who develop BP.1 There are reports of successful treatment of BP with a course of a PD1 inhibitor,9 but 2 of our patients had severe BP that led to discontinuation of immunotherapy.
Consider Prescreening
Given that development of BP often leads to cessation of therapy, identifying patients at risk prior to starting an immune checkpoint inhibitor might have clinical utility. Biopsy with DIF is the gold standard for diagnosis, but serologic testing can be a useful adjunct because enzyme-linked immunosorbent assay for BP antigen 1 and BP antigen 2 has a reported sensitivity and specificity of 87% and 98%, respectively.10 Serologic testing prior to starting therapy with an immune checkpoint inhibitor can provide a baseline for patients. A rise in titer, in conjunction with onset of a rash, might aid in earlier diagnosis, particularly because urticarial BP can be difficult to diagnose clinically.
Further study on the utility vs cost-benefit of these screening modalities is warranted. Their predictive utility might be limited, however, and positive serologic test results might have unanticipated consequences, such as hesitation in treating patients, thus leading to a delay in therapy or access to these medications.
Conclusion
The expanding use of immune checkpoint inhibitors is increasing survival in patients with metastatic melanoma and other malignancies. Adverse effects are part of the continuum of immune system stimulation, with overstimulation resulting in dermatitis; thyroiditis; pneumonitis; and less commonly hypophysitis, vitiligo, and colitis.
Rarely, immune checkpoint inhibition induces BP. Development of BP leads to discontinuation of therapy in more than half of reported cases due to lack of adequate treatment for this skin disease and its impact on quality of life. Therefore, quick diagnosis of BP in patients on immunotherapy and successful management techniques can prevent discontinuation of these lifesaving cancer therapies. For that reason, dermatologists play an important role in the management of patients on immune checkpoint inhibitors for cancer.
- Lopez AT, Khanna T, Antonov N, et al. A review of bullous pemphigoid associated with PD-1 and PD-L1 inhibitors. Int J Dermatol. 2018;57:664-669.
- Márquez-Rodas, I, Cerezuela P, Soria A, et al. Immune checkpoint inhibitors: therapeutic advances in melanoma. Ann Transl Med. 2015;3:267.
- Friedman CF, Proverbs-Singh TA, Postow MA. Treatment of the immune-related adverse effects of immune checkpoint inhibitors a review. JAMA Oncol. 2016;2:1346-1353.
- Hua C, Boussemart L, Mateus C, et al. Association of vitiligo with tumor response in patients with metastatic melanoma treated with pembrolizumab. JAMA Dermatol. 2016;152:45-51.
- Hwang SJE, Carlos G, Chou S, et al. Bullous pemphigoid, an autoantibody-mediated disease, is a novel immune-related adverse event in patients treated with anti-programmed cell death 1 antibodies. Melanoma Res. 2016;26:413-416.
- Damsky W, Kole L, Tomayko MM. Development of bullous pemphigoid during nivolumab therapy. JAAD Case Rep. 2016;2:442-444.
- Garje R, Chau JJ, Chung J, et al. Acute flare of bullous pemphigus with pembrolizumab used for treatment of metastatic urothelial cancer. J Immunother. 2018;41:42-44.
- Ito M, Hoashi T, Endo Y, et al. Atypical pemphigus developed in a patient with urothelial carcinoma treated with nivolumab. J Dermatol. 2019;46:e90-e92.
- Chen W-S, Tetzlaff MT, Diwan H, et al. Suprabasal acantholytic dermatologic toxicities associated checkpoint inhibitor therapy: a spectrum of immune reactions from paraneoplastic pemphigus-like to Grover-like lesions. J Cutan Pathol. 2018;45:764-773.
- Muglia C, Bronsnick T, Kirkorian AY, et al. Questioning the specificity and sensitivity of ELISA for bullous pemphigoid diagnosis. Cutis. 2017;99:E27-E30.
Immune checkpoint inhibitors are used for a variety of advanced malignancies, including melanoma, non–small cell lung cancer, urothelial cancer, and renal cell carcinoma. Anti–programmed cell death 1 (PD1) targeted therapies, such as pembrolizumab and nivolumab, are improving patient survival. This class of immunotherapy is revolutionary but is associated with autoimmune adverse effects. A rare but increasingly reported adverse effect of anti-PD1 therapy is bullous pemphigoid (BP), an autoimmune blistering disease directed against
High clinical suspicion, early diagnosis, and proper management of immunotherapy-related BP are imperative for keeping patients on life-prolonging treatment. We present 3 cases of BP secondary to anti-PD1 immunotherapy in patients with melanoma or non–small cell lung cancer to highlight the diagnosis and treatment of BP as well as emphasize the importance of the dermatologist in the care of patients with immunotherapy-related skin disease.
Case Reports
Patient 1
A 72-year-old woman with metastatic BRAF-mutated melanoma from an unknown primary site presented with intensely pruritic papules on the back, chest, and extremities of 4 months’ duration. She described her symptoms as insidious in onset and refractory to clobetasol ointment, oral diphenhydramine, and over-the-counter anti-itch creams. The patient had been treated with oral dabrafenib 150 mg twice daily and trametinib 2 mg/d but was switched to pembrolizumab when the disease progressed. After 8 months, she had a complete radiologic response to pembrolizumab 2 mg/kg every 3 weeks, which was discontinued in favor of observation 3 months prior to presentation to dermatology.
At the current presentation, physical examination revealed innumerable erythematous, excoriated, 2- to 4-mm, red papules diffusely scattered on the upper back, chest, abdomen, and thighs, with one 8×4-mm vesicle on the right side of the upper back (Figure 1). Discrete areas of depigmented macules, consistent with vitiligo, coalesced into patches on the legs, thighs, arms, and back. The patient was started on a 3-week oral prednisone taper for symptom relief. A hematoxylin and eosin (H&E)–stained punch biopsy of the back revealed a subepidermal split with eosinophils and a dense eosinophilic infiltrate in the dermis (Figure 2). Direct immunofluorescence (DIF) studies from a specimen adjacent to the biopsy collected for H&E staining showed linear deposition of IgA, IgG, and C3 along the dermoepidermal junction (Figure 3). Histologic findings were consistent with BP.
The patient was started on doxycycline 100 mg twice daily and clobetasol ointment 0.05% once daily to supplement the prednisone taper. At 3-week follow-up, she reported pruritus and a few erythematous macules but no new bullae. At 12 weeks, some papules persisted; however, the patient was averse to using systemic agents and decided that symptoms were adequately controlled with clobetasol ointment and oral doxycycline.
Because the patient currently remains in clinical and radiologic remission, anti-PD1 immune checkpoint inhibitors have not been restarted but remain an option for the future if disease recurs
Patient 2
An 82-year-old man with a history of stage IIC desmoplastic melanoma presented to dermatology with an intensely pruritic eruption on the legs, arms, waist, upper torso, and scalp of 3 weeks’ duration. Clobetasol ointment had provided minimal relief.
Six months prior to presenting to dermatology, the patient underwent immunotherapy with 4 cycles of ipilimumab 200 mg intravenous (IV) and nivolumab 240 mg IV every 2 weeks, receiving ipilimumab during the first cycle only because of a lack of availability at the pharmacy. He then received nivolumab 240 mg IV every 2 weeks as maintenance therapy. After the second dose of nivolumab maintenance therapy, however, he developed generalized bullae and pruritus. Dermatology was consulted during an oncology appointment, and his oncologist decided to hold nivolumab.
Physical examination revealed generalized tense and eroded bullae covering more than 50% of the body surface area and affecting the scalp, arms, legs, torso, and buttocks. Two punch biopsies were obtained. Hematoxylin and eosin staining revealed a subepidermal split with predominantly eosinophils and scattered neutrophils. Direct immunofluorescence studies showed linear deposition of IgG, IgA, and C3 along the dermoepidermal junction, consistent with BP.
The patient’s BP was difficult to control, requiring several hospital admissions for wound care, high-dose systemic steroids, and initiation of mycophenolate mofetil. After 4 months of waxing and waning symptoms, the BP was controlled with mycophenolate mofetil 1500 mg/d; clobetasol ointment 0.05%; and diphenhydramine for pruritus. Due to the prolonged recovery and severity of BP, the patient’s oncologist deemed that he was not a candidate for future immunotherapy.
Patient 3
A 68-year-old man with PD1-negative, metastatic, well-differentiated squamous cell carcinoma of the lung presented to dermatology with a pruritic rash of 3 weeks’ duration. He had been receiving nivolumab for 2 years after disease progressed on prior chemotherapies and experienced several grade 1 or grade 2 nivolumab-induced autoimmune reactions including thyroiditis, dermatitis, and nephritis, for which he was taking prednisone 5 mg/d for suppression.
Physical examination revealed psoriasiform pink plaques on the arms, chest, and legs. The differential diagnosis at the time favored psoriasiform dermatitis over lichenoid dermatitis. A punch biopsy revealed psoriasiform dermatitis. The patient was prescribed fluocinonide ointment 0.05% daily. His plaques improved with topical steroids.
The patient returned approximately 1 month later with a report of a new blistering rash on the legs. Physical examination revealed interval improvement of the psoriasiform plaques on the scalp, torso, and extremities, but tense bullae were seen on the thighs, with surrounding superficial erosions at sites of recent bullae. Punch biopsies of the skin for H&E staining and DIF showed BP.
Prednisone was increased to 50 mg/d for a 3-week taper. Doxycycline 100 mg twice daily was started. The patient’s skin disease continued to be difficult to control with therapy; nivolumab was held by his oncologist.
Comment
Immunotherapy with immune checkpoint blockade represents a successful application of immune recognition to treat metastatic cancers, including melanoma, non–small cell lung cancer, urothelial cancer, and renal cell carcinoma.
Anti-PD1 targeted therapies improve survival in solid and hematologic malignancies, with a response rate as high as 40% in melanoma.2 Although these medications can prolong survival, many are associated with loss of self-tolerance and severe autoimmunelike events that can limit therapy.3 An exception is PD1-induced vitiligo, which patient 1 developed and has been associated with a better response to therapy.4
Anti-PD1–induced BP is a newly reported adverse effect. In its early stages, BP can be difficult to differentiate from eczematous or urticarial dermatitis.5-8 Discontinuation of immunotherapy has been reported in more than 70% of patients who develop BP.1 There are reports of successful treatment of BP with a course of a PD1 inhibitor,9 but 2 of our patients had severe BP that led to discontinuation of immunotherapy.
Consider Prescreening
Given that development of BP often leads to cessation of therapy, identifying patients at risk prior to starting an immune checkpoint inhibitor might have clinical utility. Biopsy with DIF is the gold standard for diagnosis, but serologic testing can be a useful adjunct because enzyme-linked immunosorbent assay for BP antigen 1 and BP antigen 2 has a reported sensitivity and specificity of 87% and 98%, respectively.10 Serologic testing prior to starting therapy with an immune checkpoint inhibitor can provide a baseline for patients. A rise in titer, in conjunction with onset of a rash, might aid in earlier diagnosis, particularly because urticarial BP can be difficult to diagnose clinically.
Further study on the utility vs cost-benefit of these screening modalities is warranted. Their predictive utility might be limited, however, and positive serologic test results might have unanticipated consequences, such as hesitation in treating patients, thus leading to a delay in therapy or access to these medications.
Conclusion
The expanding use of immune checkpoint inhibitors is increasing survival in patients with metastatic melanoma and other malignancies. Adverse effects are part of the continuum of immune system stimulation, with overstimulation resulting in dermatitis; thyroiditis; pneumonitis; and less commonly hypophysitis, vitiligo, and colitis.
Rarely, immune checkpoint inhibition induces BP. Development of BP leads to discontinuation of therapy in more than half of reported cases due to lack of adequate treatment for this skin disease and its impact on quality of life. Therefore, quick diagnosis of BP in patients on immunotherapy and successful management techniques can prevent discontinuation of these lifesaving cancer therapies. For that reason, dermatologists play an important role in the management of patients on immune checkpoint inhibitors for cancer.
Immune checkpoint inhibitors are used for a variety of advanced malignancies, including melanoma, non–small cell lung cancer, urothelial cancer, and renal cell carcinoma. Anti–programmed cell death 1 (PD1) targeted therapies, such as pembrolizumab and nivolumab, are improving patient survival. This class of immunotherapy is revolutionary but is associated with autoimmune adverse effects. A rare but increasingly reported adverse effect of anti-PD1 therapy is bullous pemphigoid (BP), an autoimmune blistering disease directed against
High clinical suspicion, early diagnosis, and proper management of immunotherapy-related BP are imperative for keeping patients on life-prolonging treatment. We present 3 cases of BP secondary to anti-PD1 immunotherapy in patients with melanoma or non–small cell lung cancer to highlight the diagnosis and treatment of BP as well as emphasize the importance of the dermatologist in the care of patients with immunotherapy-related skin disease.
Case Reports
Patient 1
A 72-year-old woman with metastatic BRAF-mutated melanoma from an unknown primary site presented with intensely pruritic papules on the back, chest, and extremities of 4 months’ duration. She described her symptoms as insidious in onset and refractory to clobetasol ointment, oral diphenhydramine, and over-the-counter anti-itch creams. The patient had been treated with oral dabrafenib 150 mg twice daily and trametinib 2 mg/d but was switched to pembrolizumab when the disease progressed. After 8 months, she had a complete radiologic response to pembrolizumab 2 mg/kg every 3 weeks, which was discontinued in favor of observation 3 months prior to presentation to dermatology.
At the current presentation, physical examination revealed innumerable erythematous, excoriated, 2- to 4-mm, red papules diffusely scattered on the upper back, chest, abdomen, and thighs, with one 8×4-mm vesicle on the right side of the upper back (Figure 1). Discrete areas of depigmented macules, consistent with vitiligo, coalesced into patches on the legs, thighs, arms, and back. The patient was started on a 3-week oral prednisone taper for symptom relief. A hematoxylin and eosin (H&E)–stained punch biopsy of the back revealed a subepidermal split with eosinophils and a dense eosinophilic infiltrate in the dermis (Figure 2). Direct immunofluorescence (DIF) studies from a specimen adjacent to the biopsy collected for H&E staining showed linear deposition of IgA, IgG, and C3 along the dermoepidermal junction (Figure 3). Histologic findings were consistent with BP.
The patient was started on doxycycline 100 mg twice daily and clobetasol ointment 0.05% once daily to supplement the prednisone taper. At 3-week follow-up, she reported pruritus and a few erythematous macules but no new bullae. At 12 weeks, some papules persisted; however, the patient was averse to using systemic agents and decided that symptoms were adequately controlled with clobetasol ointment and oral doxycycline.
Because the patient currently remains in clinical and radiologic remission, anti-PD1 immune checkpoint inhibitors have not been restarted but remain an option for the future if disease recurs
Patient 2
An 82-year-old man with a history of stage IIC desmoplastic melanoma presented to dermatology with an intensely pruritic eruption on the legs, arms, waist, upper torso, and scalp of 3 weeks’ duration. Clobetasol ointment had provided minimal relief.
Six months prior to presenting to dermatology, the patient underwent immunotherapy with 4 cycles of ipilimumab 200 mg intravenous (IV) and nivolumab 240 mg IV every 2 weeks, receiving ipilimumab during the first cycle only because of a lack of availability at the pharmacy. He then received nivolumab 240 mg IV every 2 weeks as maintenance therapy. After the second dose of nivolumab maintenance therapy, however, he developed generalized bullae and pruritus. Dermatology was consulted during an oncology appointment, and his oncologist decided to hold nivolumab.
Physical examination revealed generalized tense and eroded bullae covering more than 50% of the body surface area and affecting the scalp, arms, legs, torso, and buttocks. Two punch biopsies were obtained. Hematoxylin and eosin staining revealed a subepidermal split with predominantly eosinophils and scattered neutrophils. Direct immunofluorescence studies showed linear deposition of IgG, IgA, and C3 along the dermoepidermal junction, consistent with BP.
The patient’s BP was difficult to control, requiring several hospital admissions for wound care, high-dose systemic steroids, and initiation of mycophenolate mofetil. After 4 months of waxing and waning symptoms, the BP was controlled with mycophenolate mofetil 1500 mg/d; clobetasol ointment 0.05%; and diphenhydramine for pruritus. Due to the prolonged recovery and severity of BP, the patient’s oncologist deemed that he was not a candidate for future immunotherapy.
Patient 3
A 68-year-old man with PD1-negative, metastatic, well-differentiated squamous cell carcinoma of the lung presented to dermatology with a pruritic rash of 3 weeks’ duration. He had been receiving nivolumab for 2 years after disease progressed on prior chemotherapies and experienced several grade 1 or grade 2 nivolumab-induced autoimmune reactions including thyroiditis, dermatitis, and nephritis, for which he was taking prednisone 5 mg/d for suppression.
Physical examination revealed psoriasiform pink plaques on the arms, chest, and legs. The differential diagnosis at the time favored psoriasiform dermatitis over lichenoid dermatitis. A punch biopsy revealed psoriasiform dermatitis. The patient was prescribed fluocinonide ointment 0.05% daily. His plaques improved with topical steroids.
The patient returned approximately 1 month later with a report of a new blistering rash on the legs. Physical examination revealed interval improvement of the psoriasiform plaques on the scalp, torso, and extremities, but tense bullae were seen on the thighs, with surrounding superficial erosions at sites of recent bullae. Punch biopsies of the skin for H&E staining and DIF showed BP.
Prednisone was increased to 50 mg/d for a 3-week taper. Doxycycline 100 mg twice daily was started. The patient’s skin disease continued to be difficult to control with therapy; nivolumab was held by his oncologist.
Comment
Immunotherapy with immune checkpoint blockade represents a successful application of immune recognition to treat metastatic cancers, including melanoma, non–small cell lung cancer, urothelial cancer, and renal cell carcinoma.
Anti-PD1 targeted therapies improve survival in solid and hematologic malignancies, with a response rate as high as 40% in melanoma.2 Although these medications can prolong survival, many are associated with loss of self-tolerance and severe autoimmunelike events that can limit therapy.3 An exception is PD1-induced vitiligo, which patient 1 developed and has been associated with a better response to therapy.4
Anti-PD1–induced BP is a newly reported adverse effect. In its early stages, BP can be difficult to differentiate from eczematous or urticarial dermatitis.5-8 Discontinuation of immunotherapy has been reported in more than 70% of patients who develop BP.1 There are reports of successful treatment of BP with a course of a PD1 inhibitor,9 but 2 of our patients had severe BP that led to discontinuation of immunotherapy.
Consider Prescreening
Given that development of BP often leads to cessation of therapy, identifying patients at risk prior to starting an immune checkpoint inhibitor might have clinical utility. Biopsy with DIF is the gold standard for diagnosis, but serologic testing can be a useful adjunct because enzyme-linked immunosorbent assay for BP antigen 1 and BP antigen 2 has a reported sensitivity and specificity of 87% and 98%, respectively.10 Serologic testing prior to starting therapy with an immune checkpoint inhibitor can provide a baseline for patients. A rise in titer, in conjunction with onset of a rash, might aid in earlier diagnosis, particularly because urticarial BP can be difficult to diagnose clinically.
Further study on the utility vs cost-benefit of these screening modalities is warranted. Their predictive utility might be limited, however, and positive serologic test results might have unanticipated consequences, such as hesitation in treating patients, thus leading to a delay in therapy or access to these medications.
Conclusion
The expanding use of immune checkpoint inhibitors is increasing survival in patients with metastatic melanoma and other malignancies. Adverse effects are part of the continuum of immune system stimulation, with overstimulation resulting in dermatitis; thyroiditis; pneumonitis; and less commonly hypophysitis, vitiligo, and colitis.
Rarely, immune checkpoint inhibition induces BP. Development of BP leads to discontinuation of therapy in more than half of reported cases due to lack of adequate treatment for this skin disease and its impact on quality of life. Therefore, quick diagnosis of BP in patients on immunotherapy and successful management techniques can prevent discontinuation of these lifesaving cancer therapies. For that reason, dermatologists play an important role in the management of patients on immune checkpoint inhibitors for cancer.
- Lopez AT, Khanna T, Antonov N, et al. A review of bullous pemphigoid associated with PD-1 and PD-L1 inhibitors. Int J Dermatol. 2018;57:664-669.
- Márquez-Rodas, I, Cerezuela P, Soria A, et al. Immune checkpoint inhibitors: therapeutic advances in melanoma. Ann Transl Med. 2015;3:267.
- Friedman CF, Proverbs-Singh TA, Postow MA. Treatment of the immune-related adverse effects of immune checkpoint inhibitors a review. JAMA Oncol. 2016;2:1346-1353.
- Hua C, Boussemart L, Mateus C, et al. Association of vitiligo with tumor response in patients with metastatic melanoma treated with pembrolizumab. JAMA Dermatol. 2016;152:45-51.
- Hwang SJE, Carlos G, Chou S, et al. Bullous pemphigoid, an autoantibody-mediated disease, is a novel immune-related adverse event in patients treated with anti-programmed cell death 1 antibodies. Melanoma Res. 2016;26:413-416.
- Damsky W, Kole L, Tomayko MM. Development of bullous pemphigoid during nivolumab therapy. JAAD Case Rep. 2016;2:442-444.
- Garje R, Chau JJ, Chung J, et al. Acute flare of bullous pemphigus with pembrolizumab used for treatment of metastatic urothelial cancer. J Immunother. 2018;41:42-44.
- Ito M, Hoashi T, Endo Y, et al. Atypical pemphigus developed in a patient with urothelial carcinoma treated with nivolumab. J Dermatol. 2019;46:e90-e92.
- Chen W-S, Tetzlaff MT, Diwan H, et al. Suprabasal acantholytic dermatologic toxicities associated checkpoint inhibitor therapy: a spectrum of immune reactions from paraneoplastic pemphigus-like to Grover-like lesions. J Cutan Pathol. 2018;45:764-773.
- Muglia C, Bronsnick T, Kirkorian AY, et al. Questioning the specificity and sensitivity of ELISA for bullous pemphigoid diagnosis. Cutis. 2017;99:E27-E30.
- Lopez AT, Khanna T, Antonov N, et al. A review of bullous pemphigoid associated with PD-1 and PD-L1 inhibitors. Int J Dermatol. 2018;57:664-669.
- Márquez-Rodas, I, Cerezuela P, Soria A, et al. Immune checkpoint inhibitors: therapeutic advances in melanoma. Ann Transl Med. 2015;3:267.
- Friedman CF, Proverbs-Singh TA, Postow MA. Treatment of the immune-related adverse effects of immune checkpoint inhibitors a review. JAMA Oncol. 2016;2:1346-1353.
- Hua C, Boussemart L, Mateus C, et al. Association of vitiligo with tumor response in patients with metastatic melanoma treated with pembrolizumab. JAMA Dermatol. 2016;152:45-51.
- Hwang SJE, Carlos G, Chou S, et al. Bullous pemphigoid, an autoantibody-mediated disease, is a novel immune-related adverse event in patients treated with anti-programmed cell death 1 antibodies. Melanoma Res. 2016;26:413-416.
- Damsky W, Kole L, Tomayko MM. Development of bullous pemphigoid during nivolumab therapy. JAAD Case Rep. 2016;2:442-444.
- Garje R, Chau JJ, Chung J, et al. Acute flare of bullous pemphigus with pembrolizumab used for treatment of metastatic urothelial cancer. J Immunother. 2018;41:42-44.
- Ito M, Hoashi T, Endo Y, et al. Atypical pemphigus developed in a patient with urothelial carcinoma treated with nivolumab. J Dermatol. 2019;46:e90-e92.
- Chen W-S, Tetzlaff MT, Diwan H, et al. Suprabasal acantholytic dermatologic toxicities associated checkpoint inhibitor therapy: a spectrum of immune reactions from paraneoplastic pemphigus-like to Grover-like lesions. J Cutan Pathol. 2018;45:764-773.
- Muglia C, Bronsnick T, Kirkorian AY, et al. Questioning the specificity and sensitivity of ELISA for bullous pemphigoid diagnosis. Cutis. 2017;99:E27-E30.
Practice Points
- Anti–programmed cell death 1 (PD1) targeted therapies improve survival in solid and hematologic malignancies but are associated with autoimmune side effects, with bullous pemphigoid (BP) being the newest reported.
- Bullous pemphigoid can develop months into immunotherapy treatment.
- Bullous pemphigoid should be on the differential diagnosis in a patient who is on an anti-PD1 immune checkpoint inhibitor and develops 1 or more of the following: pruritus, dermatitis, and vesicles.
- Early diagnosis of BP is essential for keeping patients on immunotherapy because its severity often results in temporary or permanent discontinuation of treatment.