The Meaning of Words and Why They Matter During End-of-Life Conversations

Article Type
Changed
Mon, 11/08/2021 - 12:15

Effective communication during end-of-life is crucial for health care delivery, but misinterpretation can influence how the quality of the care is rendered and perceived.

When I was a new palliative care nurse practitioner (NP), I remember my mentor telling me that communication in our field is equivalent to surgical procedures in general surgery. Conversations need to be handled with accuracy and precision, conducted in a timely fashion, and require skills that take practice to sharpen. Over the years, I learned that unlike surgery, we do not have control over how the procedure will flow. We approach patients with a blank canvas, open to receive messages that will be shared and reacted to accordingly. The ability to communicate effectively also requires compassion, which is a trait that tends to be inherent in humans and typically is not learned from textbooks but can be cultivated with training and application.

Among the barriers identified to effective communication are avoiding emotional issues and focusing on technical topics due in part to the fear of lengthy encounters, not allowing patients or families enough time to speak, and reframing instead of validating emotions.1 Many years later, I had the chance to help care for a patient whose story reminds me of how our choice of words and how our interpretation of what we are told can influence the way we care for patients and their families.

Case Presentation

Mr. P, aged 86 years, was admitted to a teaching hospital for pneumonia and heart failure exacerbation. He was treated with diuretics and antibiotics and discharged home on room air after 3 days. He returned to the hospital after 8 days, reporting labored breathing. He was found to be hypoxic, and a further workup revealed acute hypoxic respiratory failure that was likely from severe pulmonary hypertension and exacerbation of his heart failure. Left heart disease is a common cause of pulmonary hypertension, which can lead to right ventricular failure and increased mortality.2

After meeting with his pulmonologist and cardiologist, Mr. P elected for a do-not-resuscitate code status and declined to be intubated. He also refused further diagnostics and life-prolonging treatments for his conditions, including a stress test, cardiac catheterization, and a right heart catheterization. He required bilevel positive airway pressure (BPAP) support at bedtime, which he also declined. He agreed to the use of supplemental oxygen through a nasal cannula and always needed 5 liters of oxygen.

Palliative care was consulted to assist with goals of care discussion. This visit took place during the COVID-19 pandemic, but Mr. P had tested negative for the COVID virus, so the palliative care NP was able to meet with Mr. P in person. He shared his understanding of the serious nature of his condition and the likelihood of a limited life expectancy without further diagnostics and possible life-prolonging treatments. He said his goal was to go home and spend the remainder of his life with his wife. He had not been out of bed since his hospitalization except to transfer to a nearby chair with the help of his nurse due to exertional dyspnea and generalized weakness. Prior to his recent hospitalizations, he was independently ambulating and had no dyspnea when performing strenuous activities. Mr. P shared that his wife was aged in her 70s and was legally blind. He added that she did not require physical assistance, but he was unsure whether she could help him because they had not been in such a situation previously. They had a daughter who visited frequently and helped with driving them to doctors’ appointments and shopping. Mr. P shared that he wanted to go home. After explaining the option of home hospice, Mr. P decided he wanted to receive hospice services at home and asked palliative care NP to contact his daughter to let her know his wishes and to tell her more about how hospice can help with his care.

The palliative care NP met with Mr. P’s nurse and shared the outcome of her visit. His nurse asked the palliative care NP whether she was familiar with his daughter. The nurse added that she wanted the palliative care NP to know that Mr. P’s daughter was quite angry and upset with his doctors after being told about his prognosis. His doctors’ notes also indicated that Mr. P wanted them to contact his daughter regarding his condition and plans for discharge, concluding that he deferred to his daughter for medical decision making.

As Mr. P’s hospitalization took place during the COVID pandemic, a face-to-face meeting with his family was not possible. The NP spoke with Mr. P’s daughter over the phone to relay his wishes and goals for his care. Mr. P’s daughter cried at times during the conversation and asked whether his condition was really that serious. The NP allowed Mr. P’s daughter to express her sadness and allowed for periods of silence during the conversation while his daughter gathered her composure. The NP reinforced the clinical information she had been provided by the medical team. Mr. P’s daughter added that he was completely independent, not requiring supplemental oxygen and was otherwise healthy just a month prior. She also asked whether there was truly nothing else that could be done to prolong his life. The NP acknowledged her observations and explained how Mr. P’s body and organs had not been able to bounce back from the recent insults to his overall physical condition.

After being told that Mr. P’s options for treatment were limited not only by his advanced age and comorbidities, but also the limitations and goals for his care he had identified, his daughter supported her father’s decision. The palliative care NP provided her information on how home hospice assists in her father’s care at home, including symptom management, nursing visits, home equipment, family support, among others. Mr. P’s daughter also said she would relay the information to her mother and call the palliative care NP if they had additional questions or concerns.

The outcome of her visit with Mr. P and his daughter were relayed by the palliative care NP to his acute health care team through an official response to the consultation request via his electronic health record. The palliative care NP also alerted the palliative care social worker to follow-up with Mr. P, his daughter, and his acute health care team to coordinate hospice services at the time of his discharge from the hospital.

Mr. P was discharged from the hospital with home hospice services after a few days. Three weeks later, Mr. P passed away peacefully on the in-patient unit of his home hospice agency as his physical care needs became too much for his family to provide at home a few days before his death. The palliative care social worker later shared with the NP that Mr. P’s daughter shared her gratitude and satisfaction with the care he had received not only from palliative care, but also from everyone during his hospitalization.

 

 

Discussion

Key themes found in end-of-life (EOL) communication with families and caregivers include highlighting clinical deterioration, involvement in decision making, continuation of high-quality care after cessation of aggressive measures, tailoring to individuals, clarity, honesty, and use of techniques in delivery.2 Some of the techniques identified were pacing, staging, and repetition.3 Other techniques that can be beneficial include allowing for time to express one’s feelings, being comfortable with brief periods of silence, validating observations shared, among others. These themes were evident in the interactions that his health care team had with Mr. P and his daughter. With honesty and clarity, various members of the health care team repeatedly shared information regarding his clinical deterioration.

Family Influence

EOL decision-making roles within a family tend to originate from family interactional histories, familial roles as well as decision-making situations the family faces.4 The US medical and legal systems also recognize formal role assignments for surrogate decision makers.4 In the case of Mr. P, his advance directive (AD) identified his daughter as his surrogate decision maker. ADs are written statements made in advance by patients expressing their wishes and limitations for treatment as well as appointing surrogate decision makers when they become unable to decide for themselves in the future.5

During discussions about the goals for his care, Mr. P made his own medical decisions and elected to pursue a comfort-focused approach to care. His request for his health care team to reach out to his daughter was largely due to his need for assistance in explaining the complexity of his clinical condition to her and how hospice services would be helpful with his EOL care. Mr. P depended on his daughter to bring him to the hospital or to his doctors’ appointments, and she had been a major source of support for him and his wife. Contrary to the belief of some of his health care practitioners, Mr. P was not deferring his medical decisions to his daughter but rather allowing for her participation as his health care partner.

Communication between nurses and patients has been found to be challenging to both parties. Nurses express difficulties in areas that include supporting patients and families after they have had a difficult conversation with their physicians and responding to patients and family members’ emotions like anger.6 EOL care issues, such as family barriers to prognostic understanding, can interfere with psychosocial care.6 Families of patients approaching the EOL describe feeling mentally worn down and being unable to think straight, leading to feelings of being overwhelmed.7 They feel the need to be in a place where they can accept the content of difficult EOL conversations to be able to effectively engage.7

Studies have shown that family members of patients at the EOL experience stress, anxiety, fatigue and depression.8 Reactions that can be perceived as anger may not be so nor directed to the health care team. Questions raised regarding the accuracy of prognostication and treatment recommendations may not necessarily reflect concerns about the quality of care received but an exercise of advocacy in exploring other options on behalf of the patient. Allowing time for families to process the information received and react freely are necessary steps to facilitate reaching a place where they can acknowledge the information being relayed.

 

 

Communication Skills Training

Every member of the health care team should be equipped with the basic skills to have these conversations. The academic curricula for members of the health care team focuses on developing communication skills, but there has been a lack of content on palliative and EOL care.9

Due to time constraints and limited opportunities in the clinical setting, there has been an increasing use of simulation-based learning activities (SBLA) to enhance communication skills among nursing students.9 At this time, the impact of SBLA in enhancing communication competency is not fully known, but given the lack of clinical opportunities for students, this option is worth considering.9 When asked, nurses recognized the need for improved EOL communication education, training, and guidelines.10 They also felt that a multidisciplinary approach in EOL communication is beneficial. The inclusion of the End-of-Life Nursing Education Consortium (ELNEC) Core training in Bachelor of Science in Nursing programs have led to improved insight on palliative care and nurses’ role in palliative care and hospice among nursing students.11

The Palliative Care and Hospice Education and Training Act of 2017 amended the Public Health Service Act to include improving EOL training for health care providers, including talking about death and dying.12 Even though the Liaison Committee of Medical Education asked medical schools to incorporate EOL care education in the medical school curricula, there is still a lack of developmentally appropriate and supervised EOL education in medical schools.12 Training on grief also has been lacking and less likely to be mandatory among medical students and residents: Workshops are mostly conducted before they can be applied in the clinical setting.13 Meanwhile, resources are available to assist physicians in EOL conversations with patient and families, such as the Serious Illness Conversation Guide, The Conversation Project, and Stanford’s Letter Project.12

Conclusions

Palliative consultation is associated with an overall improvement in EOL care, communication, and support, according to families of deceased patients.14 It has also been shown to enhance patients’ quality of life and mood, improve documentation of resuscitation preferences, and lead to less aggressive care at the EOL, including less chemotherapy.15 Integration of palliative care in the care of patients hospitalized with acute heart failure has been associated with improved quality of life, decreased symptom burden and depressive symptoms, and increased participation in advance care planning.16

The involvement of palliative care in the care of patients and their families at EOL enhances goals of care discussions that improve understanding for everyone involved. It helps provide consistency with the message being delivered by the rest of the health care team to patients and families regarding prognosis and recommendations. Palliative care can provide an alternative when all other aggressive measures are no longer helpful and allow for the continuation of care with a shift in focus from prolonging life to promoting its quality. Furthermore, palliative care involvement for care of patients with life-limiting illness also has been found to improve symptom control, decrease hospitalizations and health care costs, and even improve mortality.17A multidisciplinary approach to palliative care EOL conversations is beneficial, but every member of the health care team should have the training, education, and skills to be ready to have these difficult conversations. These health care team members include physicians, advance practice clinicians, nurses, social workers, and chaplains, among others. Patients and families are likely to be in contact with different members of the health care team who should be able to carry out therapeutic conversations. Using validated tools and resources on communication techniques through evidence-based practice is helpful and should be encouraged. This provides a framework on how EOL conversations should be conducted in the clinical setting to augment the identified lack of training on EOL communication in schools. Repeated opportunities for its use over time will help improve the ability of clinicians to engage in effective EOL communication.

References

1. MacKenzie AR, Lasota M. Bringing life to death: the need for honest, compassionate, and effective end-of-life conversations. Am Soc Clin Oncol Educ Book. 2020;40:476-484. doi:10.1200/EDBK_279767

2. Krishnan U, Horn E. Pulmonary hypertension due to left heart disease (group 2 pulmonary hypertension) in adults. Accessed September 17, 2021. https://www.uptodate.com/contents/pulmonary-hypertension-due-to-left-heart-disease-group-2-pulmonary-hypertension-in-adults

3. Anderson RJ, Bloch S, Armstrong M, Stone PC, Low JT. Communication between healthcare professionals and relatives of patients approaching the end-of-life: a systematic review of qualitative evidence. Palliat Med. 2019;33(8):926-941. doi:10.1177/0269216319852007

4. Trees AR, Ohs JE, Murray MC. Family communication about end-of-life decisions and the enactment of the decision-maker role. Behav Sci (Basel). 2017;7(2):36. doi:10.3390/bs7020036 5. Arruda LM, Abreu KPB, Santana LBC, Sales MVC. Variables that influence the medical decision regarding advance directives and their impact on end-of-life care. Einstein (Sao Paulo). 2019;18:eRW4852. doi:10.31744/einstein_journal/2020RW4852

6. Banerjee SC, Manna R, Coyle N, et al. The implementation and evaluation of a communication skills training program for oncology nurses. Transl Behav Med. 2017;7(3):615-623. doi:10.1007/s13142-017-0473-5

7. Mitchell S, Spry JL, Hill E, Coad J, Dale J, Plunkett A. Parental experiences of end of life care decision-making for children with life-limiting conditions in the paediatric intensive care unit: a qualitative interview study. BMJ Open. 2019;9(5):e028548. doi:10.1136/bmjopen-2018-028548

8. Laryionava K, Pfeil TA, Dietrich M, Reiter-Theil S, Hiddemann W, Winkler EC. The second patient? Family members of cancer patients and their role in end-of-life decision making. BMC Palliat Care. 2018;17(1):29. doi:10.1186/s12904-018-0288-2

9. Smith MB, Macieira TGR, Bumbach MD, et al. The use of simulation to teach nursing students and clinicians palliative care and end-of-life communication: a systematic review. Am J Hosp Palliat Care. 2018;35(8):1140-1154. doi:10.1177/1049909118761386

10. Griffiths I. What are the challenges for nurses when providing end-of-life care in intensive care units? Br J Nurs. 2019;28(16):1047-1052. doi:10.12968/bjon.2019.28.16.1047

11. Li J, Smothers A, Fang W, Borland M. Undergraduate nursing students’ perception of end-of-life care education placement in the nursing curriculum. J Hosp Palliat Nurs. 2019;21(5):E12-E18. doi:10.1097/NJH.0000000000000533

12. Sutherland R. Dying well-informed: the need for better clinical educationsurrounding facilitating end-of-life conversations. Yale J Biol Med. 2019;92(4):757-764.

13. Sikstrom L, Saikaly R, Ferguson G, Mosher PJ, Bonato S, Soklaridis S. Being there: a scoping review of grief support training in medical education. PLoS One. 2019;14(11):e0224325. doi:10.1371/journal.pone.0224325

14. Yefimova M, Aslakson RA, Yang L, et al. Palliative care and end-of-life outcomes following high-risk surgery. JAMA Surg. 2020;155(2):138-146. doi:10.1001/jamasurg.2019.5083

15. Temel JS, Greer JA, Muzikansky A, et al. Early palliative care for patients with metastatic non-small-cell lung cancer. N Engl J Med. 2010;363(8):733-42. doi:10.1056/NEJMoa1000678.

16. Sidebottom AC, Jorgenson A, Richards H, Kirven J, Sillah A. Inpatient palliative care for patients with acute heart failure: outcomes from a randomized trial. J Palliat Med. 2015;18(2):134-142. doi:org/10.1089/jpm.2014.0192

17. Diop MS, Rudolph JL, Zimmerman KM, Richter MA, Skarf LM. Palliative careinterventions for patients with heart failure: a systematic review and meta-analysis. J Palliat Med. 2017;20(1):84-92. doi:10.1089/jpm.2016.0330

Article PDF
Author and Disclosure Information

Grace Cullen is a Nurse Practitioner at John D. Dingell Veterans Affairs Medical Center in Detroit, Michigan.
Correspondence: Grace Cullen ([email protected])

 

Author disclosures
The author reports no actual or potential conflicts of interest with regard to this article.

Disclaimer
The opinions expressed herein are those of the author and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies.

Issue
Federal Practitioner - 38(11)a
Publications
Topics
Page Number
497-500
Sections
Author and Disclosure Information

Grace Cullen is a Nurse Practitioner at John D. Dingell Veterans Affairs Medical Center in Detroit, Michigan.
Correspondence: Grace Cullen ([email protected])

 

Author disclosures
The author reports no actual or potential conflicts of interest with regard to this article.

Disclaimer
The opinions expressed herein are those of the author and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies.

Author and Disclosure Information

Grace Cullen is a Nurse Practitioner at John D. Dingell Veterans Affairs Medical Center in Detroit, Michigan.
Correspondence: Grace Cullen ([email protected])

 

Author disclosures
The author reports no actual or potential conflicts of interest with regard to this article.

Disclaimer
The opinions expressed herein are those of the author and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies.

Article PDF
Article PDF

Effective communication during end-of-life is crucial for health care delivery, but misinterpretation can influence how the quality of the care is rendered and perceived.

Effective communication during end-of-life is crucial for health care delivery, but misinterpretation can influence how the quality of the care is rendered and perceived.

When I was a new palliative care nurse practitioner (NP), I remember my mentor telling me that communication in our field is equivalent to surgical procedures in general surgery. Conversations need to be handled with accuracy and precision, conducted in a timely fashion, and require skills that take practice to sharpen. Over the years, I learned that unlike surgery, we do not have control over how the procedure will flow. We approach patients with a blank canvas, open to receive messages that will be shared and reacted to accordingly. The ability to communicate effectively also requires compassion, which is a trait that tends to be inherent in humans and typically is not learned from textbooks but can be cultivated with training and application.

Among the barriers identified to effective communication are avoiding emotional issues and focusing on technical topics due in part to the fear of lengthy encounters, not allowing patients or families enough time to speak, and reframing instead of validating emotions.1 Many years later, I had the chance to help care for a patient whose story reminds me of how our choice of words and how our interpretation of what we are told can influence the way we care for patients and their families.

Case Presentation

Mr. P, aged 86 years, was admitted to a teaching hospital for pneumonia and heart failure exacerbation. He was treated with diuretics and antibiotics and discharged home on room air after 3 days. He returned to the hospital after 8 days, reporting labored breathing. He was found to be hypoxic, and a further workup revealed acute hypoxic respiratory failure that was likely from severe pulmonary hypertension and exacerbation of his heart failure. Left heart disease is a common cause of pulmonary hypertension, which can lead to right ventricular failure and increased mortality.2

After meeting with his pulmonologist and cardiologist, Mr. P elected for a do-not-resuscitate code status and declined to be intubated. He also refused further diagnostics and life-prolonging treatments for his conditions, including a stress test, cardiac catheterization, and a right heart catheterization. He required bilevel positive airway pressure (BPAP) support at bedtime, which he also declined. He agreed to the use of supplemental oxygen through a nasal cannula and always needed 5 liters of oxygen.

Palliative care was consulted to assist with goals of care discussion. This visit took place during the COVID-19 pandemic, but Mr. P had tested negative for the COVID virus, so the palliative care NP was able to meet with Mr. P in person. He shared his understanding of the serious nature of his condition and the likelihood of a limited life expectancy without further diagnostics and possible life-prolonging treatments. He said his goal was to go home and spend the remainder of his life with his wife. He had not been out of bed since his hospitalization except to transfer to a nearby chair with the help of his nurse due to exertional dyspnea and generalized weakness. Prior to his recent hospitalizations, he was independently ambulating and had no dyspnea when performing strenuous activities. Mr. P shared that his wife was aged in her 70s and was legally blind. He added that she did not require physical assistance, but he was unsure whether she could help him because they had not been in such a situation previously. They had a daughter who visited frequently and helped with driving them to doctors’ appointments and shopping. Mr. P shared that he wanted to go home. After explaining the option of home hospice, Mr. P decided he wanted to receive hospice services at home and asked palliative care NP to contact his daughter to let her know his wishes and to tell her more about how hospice can help with his care.

The palliative care NP met with Mr. P’s nurse and shared the outcome of her visit. His nurse asked the palliative care NP whether she was familiar with his daughter. The nurse added that she wanted the palliative care NP to know that Mr. P’s daughter was quite angry and upset with his doctors after being told about his prognosis. His doctors’ notes also indicated that Mr. P wanted them to contact his daughter regarding his condition and plans for discharge, concluding that he deferred to his daughter for medical decision making.

As Mr. P’s hospitalization took place during the COVID pandemic, a face-to-face meeting with his family was not possible. The NP spoke with Mr. P’s daughter over the phone to relay his wishes and goals for his care. Mr. P’s daughter cried at times during the conversation and asked whether his condition was really that serious. The NP allowed Mr. P’s daughter to express her sadness and allowed for periods of silence during the conversation while his daughter gathered her composure. The NP reinforced the clinical information she had been provided by the medical team. Mr. P’s daughter added that he was completely independent, not requiring supplemental oxygen and was otherwise healthy just a month prior. She also asked whether there was truly nothing else that could be done to prolong his life. The NP acknowledged her observations and explained how Mr. P’s body and organs had not been able to bounce back from the recent insults to his overall physical condition.

After being told that Mr. P’s options for treatment were limited not only by his advanced age and comorbidities, but also the limitations and goals for his care he had identified, his daughter supported her father’s decision. The palliative care NP provided her information on how home hospice assists in her father’s care at home, including symptom management, nursing visits, home equipment, family support, among others. Mr. P’s daughter also said she would relay the information to her mother and call the palliative care NP if they had additional questions or concerns.

The outcome of her visit with Mr. P and his daughter were relayed by the palliative care NP to his acute health care team through an official response to the consultation request via his electronic health record. The palliative care NP also alerted the palliative care social worker to follow-up with Mr. P, his daughter, and his acute health care team to coordinate hospice services at the time of his discharge from the hospital.

Mr. P was discharged from the hospital with home hospice services after a few days. Three weeks later, Mr. P passed away peacefully on the in-patient unit of his home hospice agency as his physical care needs became too much for his family to provide at home a few days before his death. The palliative care social worker later shared with the NP that Mr. P’s daughter shared her gratitude and satisfaction with the care he had received not only from palliative care, but also from everyone during his hospitalization.

 

 

Discussion

Key themes found in end-of-life (EOL) communication with families and caregivers include highlighting clinical deterioration, involvement in decision making, continuation of high-quality care after cessation of aggressive measures, tailoring to individuals, clarity, honesty, and use of techniques in delivery.2 Some of the techniques identified were pacing, staging, and repetition.3 Other techniques that can be beneficial include allowing for time to express one’s feelings, being comfortable with brief periods of silence, validating observations shared, among others. These themes were evident in the interactions that his health care team had with Mr. P and his daughter. With honesty and clarity, various members of the health care team repeatedly shared information regarding his clinical deterioration.

Family Influence

EOL decision-making roles within a family tend to originate from family interactional histories, familial roles as well as decision-making situations the family faces.4 The US medical and legal systems also recognize formal role assignments for surrogate decision makers.4 In the case of Mr. P, his advance directive (AD) identified his daughter as his surrogate decision maker. ADs are written statements made in advance by patients expressing their wishes and limitations for treatment as well as appointing surrogate decision makers when they become unable to decide for themselves in the future.5

During discussions about the goals for his care, Mr. P made his own medical decisions and elected to pursue a comfort-focused approach to care. His request for his health care team to reach out to his daughter was largely due to his need for assistance in explaining the complexity of his clinical condition to her and how hospice services would be helpful with his EOL care. Mr. P depended on his daughter to bring him to the hospital or to his doctors’ appointments, and she had been a major source of support for him and his wife. Contrary to the belief of some of his health care practitioners, Mr. P was not deferring his medical decisions to his daughter but rather allowing for her participation as his health care partner.

Communication between nurses and patients has been found to be challenging to both parties. Nurses express difficulties in areas that include supporting patients and families after they have had a difficult conversation with their physicians and responding to patients and family members’ emotions like anger.6 EOL care issues, such as family barriers to prognostic understanding, can interfere with psychosocial care.6 Families of patients approaching the EOL describe feeling mentally worn down and being unable to think straight, leading to feelings of being overwhelmed.7 They feel the need to be in a place where they can accept the content of difficult EOL conversations to be able to effectively engage.7

Studies have shown that family members of patients at the EOL experience stress, anxiety, fatigue and depression.8 Reactions that can be perceived as anger may not be so nor directed to the health care team. Questions raised regarding the accuracy of prognostication and treatment recommendations may not necessarily reflect concerns about the quality of care received but an exercise of advocacy in exploring other options on behalf of the patient. Allowing time for families to process the information received and react freely are necessary steps to facilitate reaching a place where they can acknowledge the information being relayed.

 

 

Communication Skills Training

Every member of the health care team should be equipped with the basic skills to have these conversations. The academic curricula for members of the health care team focuses on developing communication skills, but there has been a lack of content on palliative and EOL care.9

Due to time constraints and limited opportunities in the clinical setting, there has been an increasing use of simulation-based learning activities (SBLA) to enhance communication skills among nursing students.9 At this time, the impact of SBLA in enhancing communication competency is not fully known, but given the lack of clinical opportunities for students, this option is worth considering.9 When asked, nurses recognized the need for improved EOL communication education, training, and guidelines.10 They also felt that a multidisciplinary approach in EOL communication is beneficial. The inclusion of the End-of-Life Nursing Education Consortium (ELNEC) Core training in Bachelor of Science in Nursing programs have led to improved insight on palliative care and nurses’ role in palliative care and hospice among nursing students.11

The Palliative Care and Hospice Education and Training Act of 2017 amended the Public Health Service Act to include improving EOL training for health care providers, including talking about death and dying.12 Even though the Liaison Committee of Medical Education asked medical schools to incorporate EOL care education in the medical school curricula, there is still a lack of developmentally appropriate and supervised EOL education in medical schools.12 Training on grief also has been lacking and less likely to be mandatory among medical students and residents: Workshops are mostly conducted before they can be applied in the clinical setting.13 Meanwhile, resources are available to assist physicians in EOL conversations with patient and families, such as the Serious Illness Conversation Guide, The Conversation Project, and Stanford’s Letter Project.12

Conclusions

Palliative consultation is associated with an overall improvement in EOL care, communication, and support, according to families of deceased patients.14 It has also been shown to enhance patients’ quality of life and mood, improve documentation of resuscitation preferences, and lead to less aggressive care at the EOL, including less chemotherapy.15 Integration of palliative care in the care of patients hospitalized with acute heart failure has been associated with improved quality of life, decreased symptom burden and depressive symptoms, and increased participation in advance care planning.16

The involvement of palliative care in the care of patients and their families at EOL enhances goals of care discussions that improve understanding for everyone involved. It helps provide consistency with the message being delivered by the rest of the health care team to patients and families regarding prognosis and recommendations. Palliative care can provide an alternative when all other aggressive measures are no longer helpful and allow for the continuation of care with a shift in focus from prolonging life to promoting its quality. Furthermore, palliative care involvement for care of patients with life-limiting illness also has been found to improve symptom control, decrease hospitalizations and health care costs, and even improve mortality.17A multidisciplinary approach to palliative care EOL conversations is beneficial, but every member of the health care team should have the training, education, and skills to be ready to have these difficult conversations. These health care team members include physicians, advance practice clinicians, nurses, social workers, and chaplains, among others. Patients and families are likely to be in contact with different members of the health care team who should be able to carry out therapeutic conversations. Using validated tools and resources on communication techniques through evidence-based practice is helpful and should be encouraged. This provides a framework on how EOL conversations should be conducted in the clinical setting to augment the identified lack of training on EOL communication in schools. Repeated opportunities for its use over time will help improve the ability of clinicians to engage in effective EOL communication.

When I was a new palliative care nurse practitioner (NP), I remember my mentor telling me that communication in our field is equivalent to surgical procedures in general surgery. Conversations need to be handled with accuracy and precision, conducted in a timely fashion, and require skills that take practice to sharpen. Over the years, I learned that unlike surgery, we do not have control over how the procedure will flow. We approach patients with a blank canvas, open to receive messages that will be shared and reacted to accordingly. The ability to communicate effectively also requires compassion, which is a trait that tends to be inherent in humans and typically is not learned from textbooks but can be cultivated with training and application.

Among the barriers identified to effective communication are avoiding emotional issues and focusing on technical topics due in part to the fear of lengthy encounters, not allowing patients or families enough time to speak, and reframing instead of validating emotions.1 Many years later, I had the chance to help care for a patient whose story reminds me of how our choice of words and how our interpretation of what we are told can influence the way we care for patients and their families.

Case Presentation

Mr. P, aged 86 years, was admitted to a teaching hospital for pneumonia and heart failure exacerbation. He was treated with diuretics and antibiotics and discharged home on room air after 3 days. He returned to the hospital after 8 days, reporting labored breathing. He was found to be hypoxic, and a further workup revealed acute hypoxic respiratory failure that was likely from severe pulmonary hypertension and exacerbation of his heart failure. Left heart disease is a common cause of pulmonary hypertension, which can lead to right ventricular failure and increased mortality.2

After meeting with his pulmonologist and cardiologist, Mr. P elected for a do-not-resuscitate code status and declined to be intubated. He also refused further diagnostics and life-prolonging treatments for his conditions, including a stress test, cardiac catheterization, and a right heart catheterization. He required bilevel positive airway pressure (BPAP) support at bedtime, which he also declined. He agreed to the use of supplemental oxygen through a nasal cannula and always needed 5 liters of oxygen.

Palliative care was consulted to assist with goals of care discussion. This visit took place during the COVID-19 pandemic, but Mr. P had tested negative for the COVID virus, so the palliative care NP was able to meet with Mr. P in person. He shared his understanding of the serious nature of his condition and the likelihood of a limited life expectancy without further diagnostics and possible life-prolonging treatments. He said his goal was to go home and spend the remainder of his life with his wife. He had not been out of bed since his hospitalization except to transfer to a nearby chair with the help of his nurse due to exertional dyspnea and generalized weakness. Prior to his recent hospitalizations, he was independently ambulating and had no dyspnea when performing strenuous activities. Mr. P shared that his wife was aged in her 70s and was legally blind. He added that she did not require physical assistance, but he was unsure whether she could help him because they had not been in such a situation previously. They had a daughter who visited frequently and helped with driving them to doctors’ appointments and shopping. Mr. P shared that he wanted to go home. After explaining the option of home hospice, Mr. P decided he wanted to receive hospice services at home and asked palliative care NP to contact his daughter to let her know his wishes and to tell her more about how hospice can help with his care.

The palliative care NP met with Mr. P’s nurse and shared the outcome of her visit. His nurse asked the palliative care NP whether she was familiar with his daughter. The nurse added that she wanted the palliative care NP to know that Mr. P’s daughter was quite angry and upset with his doctors after being told about his prognosis. His doctors’ notes also indicated that Mr. P wanted them to contact his daughter regarding his condition and plans for discharge, concluding that he deferred to his daughter for medical decision making.

As Mr. P’s hospitalization took place during the COVID pandemic, a face-to-face meeting with his family was not possible. The NP spoke with Mr. P’s daughter over the phone to relay his wishes and goals for his care. Mr. P’s daughter cried at times during the conversation and asked whether his condition was really that serious. The NP allowed Mr. P’s daughter to express her sadness and allowed for periods of silence during the conversation while his daughter gathered her composure. The NP reinforced the clinical information she had been provided by the medical team. Mr. P’s daughter added that he was completely independent, not requiring supplemental oxygen and was otherwise healthy just a month prior. She also asked whether there was truly nothing else that could be done to prolong his life. The NP acknowledged her observations and explained how Mr. P’s body and organs had not been able to bounce back from the recent insults to his overall physical condition.

After being told that Mr. P’s options for treatment were limited not only by his advanced age and comorbidities, but also the limitations and goals for his care he had identified, his daughter supported her father’s decision. The palliative care NP provided her information on how home hospice assists in her father’s care at home, including symptom management, nursing visits, home equipment, family support, among others. Mr. P’s daughter also said she would relay the information to her mother and call the palliative care NP if they had additional questions or concerns.

The outcome of her visit with Mr. P and his daughter were relayed by the palliative care NP to his acute health care team through an official response to the consultation request via his electronic health record. The palliative care NP also alerted the palliative care social worker to follow-up with Mr. P, his daughter, and his acute health care team to coordinate hospice services at the time of his discharge from the hospital.

Mr. P was discharged from the hospital with home hospice services after a few days. Three weeks later, Mr. P passed away peacefully on the in-patient unit of his home hospice agency as his physical care needs became too much for his family to provide at home a few days before his death. The palliative care social worker later shared with the NP that Mr. P’s daughter shared her gratitude and satisfaction with the care he had received not only from palliative care, but also from everyone during his hospitalization.

 

 

Discussion

Key themes found in end-of-life (EOL) communication with families and caregivers include highlighting clinical deterioration, involvement in decision making, continuation of high-quality care after cessation of aggressive measures, tailoring to individuals, clarity, honesty, and use of techniques in delivery.2 Some of the techniques identified were pacing, staging, and repetition.3 Other techniques that can be beneficial include allowing for time to express one’s feelings, being comfortable with brief periods of silence, validating observations shared, among others. These themes were evident in the interactions that his health care team had with Mr. P and his daughter. With honesty and clarity, various members of the health care team repeatedly shared information regarding his clinical deterioration.

Family Influence

EOL decision-making roles within a family tend to originate from family interactional histories, familial roles as well as decision-making situations the family faces.4 The US medical and legal systems also recognize formal role assignments for surrogate decision makers.4 In the case of Mr. P, his advance directive (AD) identified his daughter as his surrogate decision maker. ADs are written statements made in advance by patients expressing their wishes and limitations for treatment as well as appointing surrogate decision makers when they become unable to decide for themselves in the future.5

During discussions about the goals for his care, Mr. P made his own medical decisions and elected to pursue a comfort-focused approach to care. His request for his health care team to reach out to his daughter was largely due to his need for assistance in explaining the complexity of his clinical condition to her and how hospice services would be helpful with his EOL care. Mr. P depended on his daughter to bring him to the hospital or to his doctors’ appointments, and she had been a major source of support for him and his wife. Contrary to the belief of some of his health care practitioners, Mr. P was not deferring his medical decisions to his daughter but rather allowing for her participation as his health care partner.

Communication between nurses and patients has been found to be challenging to both parties. Nurses express difficulties in areas that include supporting patients and families after they have had a difficult conversation with their physicians and responding to patients and family members’ emotions like anger.6 EOL care issues, such as family barriers to prognostic understanding, can interfere with psychosocial care.6 Families of patients approaching the EOL describe feeling mentally worn down and being unable to think straight, leading to feelings of being overwhelmed.7 They feel the need to be in a place where they can accept the content of difficult EOL conversations to be able to effectively engage.7

Studies have shown that family members of patients at the EOL experience stress, anxiety, fatigue and depression.8 Reactions that can be perceived as anger may not be so nor directed to the health care team. Questions raised regarding the accuracy of prognostication and treatment recommendations may not necessarily reflect concerns about the quality of care received but an exercise of advocacy in exploring other options on behalf of the patient. Allowing time for families to process the information received and react freely are necessary steps to facilitate reaching a place where they can acknowledge the information being relayed.

 

 

Communication Skills Training

Every member of the health care team should be equipped with the basic skills to have these conversations. The academic curricula for members of the health care team focuses on developing communication skills, but there has been a lack of content on palliative and EOL care.9

Due to time constraints and limited opportunities in the clinical setting, there has been an increasing use of simulation-based learning activities (SBLA) to enhance communication skills among nursing students.9 At this time, the impact of SBLA in enhancing communication competency is not fully known, but given the lack of clinical opportunities for students, this option is worth considering.9 When asked, nurses recognized the need for improved EOL communication education, training, and guidelines.10 They also felt that a multidisciplinary approach in EOL communication is beneficial. The inclusion of the End-of-Life Nursing Education Consortium (ELNEC) Core training in Bachelor of Science in Nursing programs have led to improved insight on palliative care and nurses’ role in palliative care and hospice among nursing students.11

The Palliative Care and Hospice Education and Training Act of 2017 amended the Public Health Service Act to include improving EOL training for health care providers, including talking about death and dying.12 Even though the Liaison Committee of Medical Education asked medical schools to incorporate EOL care education in the medical school curricula, there is still a lack of developmentally appropriate and supervised EOL education in medical schools.12 Training on grief also has been lacking and less likely to be mandatory among medical students and residents: Workshops are mostly conducted before they can be applied in the clinical setting.13 Meanwhile, resources are available to assist physicians in EOL conversations with patient and families, such as the Serious Illness Conversation Guide, The Conversation Project, and Stanford’s Letter Project.12

Conclusions

Palliative consultation is associated with an overall improvement in EOL care, communication, and support, according to families of deceased patients.14 It has also been shown to enhance patients’ quality of life and mood, improve documentation of resuscitation preferences, and lead to less aggressive care at the EOL, including less chemotherapy.15 Integration of palliative care in the care of patients hospitalized with acute heart failure has been associated with improved quality of life, decreased symptom burden and depressive symptoms, and increased participation in advance care planning.16

The involvement of palliative care in the care of patients and their families at EOL enhances goals of care discussions that improve understanding for everyone involved. It helps provide consistency with the message being delivered by the rest of the health care team to patients and families regarding prognosis and recommendations. Palliative care can provide an alternative when all other aggressive measures are no longer helpful and allow for the continuation of care with a shift in focus from prolonging life to promoting its quality. Furthermore, palliative care involvement for care of patients with life-limiting illness also has been found to improve symptom control, decrease hospitalizations and health care costs, and even improve mortality.17A multidisciplinary approach to palliative care EOL conversations is beneficial, but every member of the health care team should have the training, education, and skills to be ready to have these difficult conversations. These health care team members include physicians, advance practice clinicians, nurses, social workers, and chaplains, among others. Patients and families are likely to be in contact with different members of the health care team who should be able to carry out therapeutic conversations. Using validated tools and resources on communication techniques through evidence-based practice is helpful and should be encouraged. This provides a framework on how EOL conversations should be conducted in the clinical setting to augment the identified lack of training on EOL communication in schools. Repeated opportunities for its use over time will help improve the ability of clinicians to engage in effective EOL communication.

References

1. MacKenzie AR, Lasota M. Bringing life to death: the need for honest, compassionate, and effective end-of-life conversations. Am Soc Clin Oncol Educ Book. 2020;40:476-484. doi:10.1200/EDBK_279767

2. Krishnan U, Horn E. Pulmonary hypertension due to left heart disease (group 2 pulmonary hypertension) in adults. Accessed September 17, 2021. https://www.uptodate.com/contents/pulmonary-hypertension-due-to-left-heart-disease-group-2-pulmonary-hypertension-in-adults

3. Anderson RJ, Bloch S, Armstrong M, Stone PC, Low JT. Communication between healthcare professionals and relatives of patients approaching the end-of-life: a systematic review of qualitative evidence. Palliat Med. 2019;33(8):926-941. doi:10.1177/0269216319852007

4. Trees AR, Ohs JE, Murray MC. Family communication about end-of-life decisions and the enactment of the decision-maker role. Behav Sci (Basel). 2017;7(2):36. doi:10.3390/bs7020036 5. Arruda LM, Abreu KPB, Santana LBC, Sales MVC. Variables that influence the medical decision regarding advance directives and their impact on end-of-life care. Einstein (Sao Paulo). 2019;18:eRW4852. doi:10.31744/einstein_journal/2020RW4852

6. Banerjee SC, Manna R, Coyle N, et al. The implementation and evaluation of a communication skills training program for oncology nurses. Transl Behav Med. 2017;7(3):615-623. doi:10.1007/s13142-017-0473-5

7. Mitchell S, Spry JL, Hill E, Coad J, Dale J, Plunkett A. Parental experiences of end of life care decision-making for children with life-limiting conditions in the paediatric intensive care unit: a qualitative interview study. BMJ Open. 2019;9(5):e028548. doi:10.1136/bmjopen-2018-028548

8. Laryionava K, Pfeil TA, Dietrich M, Reiter-Theil S, Hiddemann W, Winkler EC. The second patient? Family members of cancer patients and their role in end-of-life decision making. BMC Palliat Care. 2018;17(1):29. doi:10.1186/s12904-018-0288-2

9. Smith MB, Macieira TGR, Bumbach MD, et al. The use of simulation to teach nursing students and clinicians palliative care and end-of-life communication: a systematic review. Am J Hosp Palliat Care. 2018;35(8):1140-1154. doi:10.1177/1049909118761386

10. Griffiths I. What are the challenges for nurses when providing end-of-life care in intensive care units? Br J Nurs. 2019;28(16):1047-1052. doi:10.12968/bjon.2019.28.16.1047

11. Li J, Smothers A, Fang W, Borland M. Undergraduate nursing students’ perception of end-of-life care education placement in the nursing curriculum. J Hosp Palliat Nurs. 2019;21(5):E12-E18. doi:10.1097/NJH.0000000000000533

12. Sutherland R. Dying well-informed: the need for better clinical educationsurrounding facilitating end-of-life conversations. Yale J Biol Med. 2019;92(4):757-764.

13. Sikstrom L, Saikaly R, Ferguson G, Mosher PJ, Bonato S, Soklaridis S. Being there: a scoping review of grief support training in medical education. PLoS One. 2019;14(11):e0224325. doi:10.1371/journal.pone.0224325

14. Yefimova M, Aslakson RA, Yang L, et al. Palliative care and end-of-life outcomes following high-risk surgery. JAMA Surg. 2020;155(2):138-146. doi:10.1001/jamasurg.2019.5083

15. Temel JS, Greer JA, Muzikansky A, et al. Early palliative care for patients with metastatic non-small-cell lung cancer. N Engl J Med. 2010;363(8):733-42. doi:10.1056/NEJMoa1000678.

16. Sidebottom AC, Jorgenson A, Richards H, Kirven J, Sillah A. Inpatient palliative care for patients with acute heart failure: outcomes from a randomized trial. J Palliat Med. 2015;18(2):134-142. doi:org/10.1089/jpm.2014.0192

17. Diop MS, Rudolph JL, Zimmerman KM, Richter MA, Skarf LM. Palliative careinterventions for patients with heart failure: a systematic review and meta-analysis. J Palliat Med. 2017;20(1):84-92. doi:10.1089/jpm.2016.0330

References

1. MacKenzie AR, Lasota M. Bringing life to death: the need for honest, compassionate, and effective end-of-life conversations. Am Soc Clin Oncol Educ Book. 2020;40:476-484. doi:10.1200/EDBK_279767

2. Krishnan U, Horn E. Pulmonary hypertension due to left heart disease (group 2 pulmonary hypertension) in adults. Accessed September 17, 2021. https://www.uptodate.com/contents/pulmonary-hypertension-due-to-left-heart-disease-group-2-pulmonary-hypertension-in-adults

3. Anderson RJ, Bloch S, Armstrong M, Stone PC, Low JT. Communication between healthcare professionals and relatives of patients approaching the end-of-life: a systematic review of qualitative evidence. Palliat Med. 2019;33(8):926-941. doi:10.1177/0269216319852007

4. Trees AR, Ohs JE, Murray MC. Family communication about end-of-life decisions and the enactment of the decision-maker role. Behav Sci (Basel). 2017;7(2):36. doi:10.3390/bs7020036 5. Arruda LM, Abreu KPB, Santana LBC, Sales MVC. Variables that influence the medical decision regarding advance directives and their impact on end-of-life care. Einstein (Sao Paulo). 2019;18:eRW4852. doi:10.31744/einstein_journal/2020RW4852

6. Banerjee SC, Manna R, Coyle N, et al. The implementation and evaluation of a communication skills training program for oncology nurses. Transl Behav Med. 2017;7(3):615-623. doi:10.1007/s13142-017-0473-5

7. Mitchell S, Spry JL, Hill E, Coad J, Dale J, Plunkett A. Parental experiences of end of life care decision-making for children with life-limiting conditions in the paediatric intensive care unit: a qualitative interview study. BMJ Open. 2019;9(5):e028548. doi:10.1136/bmjopen-2018-028548

8. Laryionava K, Pfeil TA, Dietrich M, Reiter-Theil S, Hiddemann W, Winkler EC. The second patient? Family members of cancer patients and their role in end-of-life decision making. BMC Palliat Care. 2018;17(1):29. doi:10.1186/s12904-018-0288-2

9. Smith MB, Macieira TGR, Bumbach MD, et al. The use of simulation to teach nursing students and clinicians palliative care and end-of-life communication: a systematic review. Am J Hosp Palliat Care. 2018;35(8):1140-1154. doi:10.1177/1049909118761386

10. Griffiths I. What are the challenges for nurses when providing end-of-life care in intensive care units? Br J Nurs. 2019;28(16):1047-1052. doi:10.12968/bjon.2019.28.16.1047

11. Li J, Smothers A, Fang W, Borland M. Undergraduate nursing students’ perception of end-of-life care education placement in the nursing curriculum. J Hosp Palliat Nurs. 2019;21(5):E12-E18. doi:10.1097/NJH.0000000000000533

12. Sutherland R. Dying well-informed: the need for better clinical educationsurrounding facilitating end-of-life conversations. Yale J Biol Med. 2019;92(4):757-764.

13. Sikstrom L, Saikaly R, Ferguson G, Mosher PJ, Bonato S, Soklaridis S. Being there: a scoping review of grief support training in medical education. PLoS One. 2019;14(11):e0224325. doi:10.1371/journal.pone.0224325

14. Yefimova M, Aslakson RA, Yang L, et al. Palliative care and end-of-life outcomes following high-risk surgery. JAMA Surg. 2020;155(2):138-146. doi:10.1001/jamasurg.2019.5083

15. Temel JS, Greer JA, Muzikansky A, et al. Early palliative care for patients with metastatic non-small-cell lung cancer. N Engl J Med. 2010;363(8):733-42. doi:10.1056/NEJMoa1000678.

16. Sidebottom AC, Jorgenson A, Richards H, Kirven J, Sillah A. Inpatient palliative care for patients with acute heart failure: outcomes from a randomized trial. J Palliat Med. 2015;18(2):134-142. doi:org/10.1089/jpm.2014.0192

17. Diop MS, Rudolph JL, Zimmerman KM, Richter MA, Skarf LM. Palliative careinterventions for patients with heart failure: a systematic review and meta-analysis. J Palliat Med. 2017;20(1):84-92. doi:10.1089/jpm.2016.0330

Issue
Federal Practitioner - 38(11)a
Issue
Federal Practitioner - 38(11)a
Page Number
497-500
Page Number
497-500
Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Pembrolizumab-Induced Type 1 Diabetes in a 95-Year-Old Veteran With Metastatic Melanoma

Article Type
Changed
Tue, 05/03/2022 - 15:03
Low C-peptide levels should prompt a high suspicion for immune checkpoint inhibitor-induced type 1 diabetes, and initiation of insulin therapy should be strongly considered.

Immune checkpoint inhibitors (CPIs) have revolutionized cancer therapy and improved the prognosis for a variety of advanced solid tumors and Hodgkin lymphoma, but evidence is growing regarding severe endocrine disturbances.1,2 CPIs block inhibitory molecules on activated T cells to increase tumor cell destruction but also can breach normal tolerance, resulting in a spectrum of immune-related adverse events (irAE).1,2 Programmed cell death-1 (PD-1) inhibitors are one type of CPIs. Pembrolizumab is a humanized monoclonal antibody that targets the PD-1 checkpoint pathway and is approved for the treatment of malignant melanoma and non-small cell lung cancer.3,4 When the PD-1 checkpoint pathway is inhibited, T cells targeting cancer are activated, as are autoreactive T cells, such as those regulating pancreatic islet cell survival, which can lead to type 1 diabetes mellitus (T1DM).5

Case Presentation

A 95-year-old male veteran with long-standing, stable prediabetes was treated with pembrolizumab for stage 4 melanoma. Four months after treatment initiation and 3 weeks after completion of his sixth treatment cycle of pembrolizumab (2 mg/kg every 3 weeks), he presented for surveillance positron emission tomography (PET) and was incidentally found to have a serum glucose of 423 mg/dL. Hypothesis-driven history taking revealed polyuria, polydipsia, and a 12-lb weight loss during the previous 3 months. The patient reported no abdominal pain, nausea, or vomiting. He showed no evidence of pancreatic metastases on recent imaging. His family history was notable for a daughter with T1DM diagnosed at a young age.

On examination, the patient’s vital signs were normal aside from a blood pressure of 80/40 mm Hg. His body mass index was 30. He was alert and oriented with comfortable respirations and no Kussmaul breathing. He exhibited dry mucous membranes and poor skin turgor. Laboratory studies revealed 135 mmol/L sodium (reference, 135-145), 4.6 mmol/L potassium (reference, 3.6-5.2), 100 mmol/L chloride (reference, 99-106), bicarbonate of 26.5 mmol/L (reference, 23-29), serum blood urea nitrogen 27 mg/dL (reference, 6-24), 1.06 mg/dL creatinine (reference, 0.74-1.35), and 423 mg/dL glucose (reference, 70-100), with negative urine ketones. Further studies demonstrated 462 µmol/L fructosamine (reference, 190-270), correlating with hemoglobin A1c (HbA1c) close to 11.0% (HbA1c was drawn on admission but cancelled by the laboratory for unknown reasons).6,7 Later, an inappropriately low C-peptide level of 0.56 ng/mL (reference, 0.8-3.85) and a negative antiglutamic acid decarboxylase (GAD) antibody titer resulted. The patient was given IV hydration and admitted to the hospital. With input from endocrinology, the patient was started on 0.3 units per kg of body weight basal-prandial insulin therapy. Pembrolizumab was held. Six weeks after discharge, his HbA1c was 7.2%, and C-peptide improved to 1.95 ng/mL and plasma glucose 116 mg/dL. After shared decision making with his health care team, the patient decided against restarting pembrolizumab. The patient reported that his functional status was preserved, and he preferred to take fewer medications at his advanced age. He died comfortably 6 months after this presentation from complications of metastatic melanoma.

Dicussion

Immunotherapy is now an integral part of cancer treatment and can result in endocrine disturbances.1,2 Life-threatening irAEs are rare and may mimic more common conditions; thus, there is growing recognition of the need to educate health care professionals in appropriate screening and management of these conditions. CPI-induced T1DM is an uncommon but clinically significant event with an incidence of 0.4 to 1.27% and a median onset of 20 weeks after initiation of therapy (range, 1-228 weeks).8-12In case seriesfrom 3 academic centers, 59 to81% of patients with CPI-induced T1DM presented with diabetic ketoacidosis (DKA), and only 40 to 71% of patients were autoantibody positive.13-16 These patients are older than those presenting with classic T1DM, often require intensive care unit admission, and nearly invariably require exogenous insulin injections for metabolic control.13-16

Based on the later age of onset of cancers that may be treated with CPI, patients with CPI-induced T1DM may be misdiagnosed with T2DM or hyperglycemia from other causes, such as medications or acute illness in the outpatient setting, risking suboptimal treatment.

Given the infrequent incidence and lack of controlled trials, screening and treatment recommendations for CPI-induced T1DM are based on principles derived from case series and expert opinion. Development of polyuria, polydipsia, weight loss, nausea, and/or vomiting should prompt investigation for possible development or worsening of hyperglycemia, suggestive of development of T1DM.17 American Society of Clinical Oncology (ASCO) guidelines recommend that serum glucose be assessed at baseline and with each treatment cycle during induction for 12 weeks, then every 3 to 6 weeks thereafter.17 There is no reported association between the number of CPI treatments and the development of DM.8,9 Following our patient’s fifth pembrolizumab cycle, a random glucose reading was noted to be 186 mg/dL (Figure 1). Under the ASCO guidelines, ideally the patient would have received close clinical follow-up given the striking increase in plasma glucose compared with prior baseline lower values and perhaps been further evaluated with an anti-GAD antibody titer to screen for T1DM.17

Glycemic Markers During Pembrolizumab Treatmenta Figure


This patient's case adds to the published reports of CPI-induced T1DM without DKA and represents the oldest patient experiencing this irAE in the literature.13-16 The degree of elevation of his initial fructosamine, which is comparable to an average plasma glucose of approximately 270 mg/dL, belied the rapid rate of rise of his recent plasma glucose. Given the trajectory of glycemic markers and symptoms, one could certainly be concerned about imminent decompensation to DKA. However, fortuitous point-of-care glucose reading prior to surveillance PET resulted in a new critical diagnosis and initiation of treatment.

 

 



Assessing the need for inpatient evaluation includes obtaining urine ketones and acid-base status as screening for DKA.17 Antibodies and C-peptide can be sent to support diagnosis of new onset T1DM, although the initiation of therapy should not be delayed for these results.17 As noted before, many of these patients also are antibody negative.13-16 Low C-peptide levels should prompt a high suspicion for CPI-induced T1DM, and initiation of insulin therapy should be strongly considered.17 In a case series of 27 patients, 85% exhibited a rapid loss of β-cell function, evidenced by the acute progression to hyperglycemia and low or undetectable levels of C-peptide at diagnosis.9 Likewise, our patient had a low C-peptide level and negative anti-GAD antibody titer but was treated before these results were available. Inpatient admission for close glycemic monitoring may be reasonable; several cases reported prompt diagnosis and avoidance of DKA in this setting.17

In contrast to other irAEs, there is no available evidence that high-dose corticosteroids alter the course of pembrolizumab-induced T2DM.18 Depending on the degree of hyperglycemia, endocrinology consultation and insulin treatment are appropriate where the diagnosis of T1DM is suspected even without evidence of DKA.17 For patients with T2DM, there may be a positive synergistic effect of metformin in combination with CPIs in tumor control.19 Our patient’s C-peptide improved with insulin treatment, consistent with correction of glucose toxicity and a honeymoon period in his course. However, in patients reported with pembrolizumab-induced T1DM, insulin requirement for treatment generally persists despite cessation of pembrolizumab therapy.13-16

Conclusions

Pembrolizumab-induced T1DM is a rare, but potentially life-threatening irAE. The acute risk of DKA requires early recognition and prompt treatment of patients taking CPIs. More than 90% of primary care physicians (PCPs) fulfill general medical care roles for patients with cancer; therefore, they play an essential role in evaluating symptoms during therapy.20 Further studies evaluating the role of PCPs and outcomes when PCPs are involved in oncologic care should be conducted.

Figure of Letter

With increased index of suspicion, this clinical scenario presents an opportunity for PCPs that may help reduce irAE-associated morbidity and mortality of patients on CPIs, like pembrolizumab. Figure 2 illustrates an example addendum that can be used to alert and tag a PCP of a mutual patient after initiation of CPI therapy. Determining the optimal interface between PCPs, oncologists, and endocrinologists in delivering and coordinating high-quality cancer care in the setting of immunotherapy is an important area for ongoing quality improvement.

Acknowledgment

The authors thank all the staff and health care professionals at VA Greater Los Angeles Healthcare System who were involved in the care of this patient.

References

1. Puzanov I, Diab A, Abdallah K, et al; Society for Immunotherapy of Cancer Toxicity Management Working Group. Managing toxicities associated with immune checkpoint inhibitors: consensus recommendations from the Society for Immunotherapy of Cancer (SITC) Toxicity Management Working Group. J Immunother Cancer. 2017;5(1):95. doi:10.1186/s40425-017-0300-z

2. Villa NM, Farahmand A, Du L, et al. Endocrinopathies with use of cancer immunotherapies. Clin Endocrinol (Oxf). 2018;88(2):327-332. doi:10.1111/cen.13483

3. Schachter J, Ribas A, Long GV, et al. Pembrolizumab versus ipilimumab for advanced melanoma: final overall survival results of a multicentre, randomised, open-label phase 3 study (KEYNOTE-006). Lancet. 2017;390(10105):1853-1862. doi:10.1016/S0140-6736(17)31601-X

4. Garon EB, Hellmann MD, Rizvi NA, et al. Five-year overall survival for patients with advanced non-small-cell lung cancer treated with pembrolizumab: results from the phase I KEYNOTE-001 Study. J Clin Oncol. 2019;37(28):2518-2527. doi:10.1200/JCO.19.00934

5. Ribas A. Tumor immunotherapy directed at PD-1. N Engl J Med. 2012;366(26):2517-2519. doi:10.1056/NEJMe1205943

6. Malmstrom H, Walldius G, Grill V, Jungner I, Gudbjomsdottir S, Hammar N. Frustosamine is a useful indicator of hyperglycemia and glucose control in clinical and epidemiological studies- cross-sectional and longitudinal experience from the AMORIS cohort. PLoS One. 2014;9(10):e111463. doi:10.1371/journal.pone.0111463

7. Skinner S, Diaw M, Mbaye MN, et al. Evaluation of agreement between hemoglobin A1c, fasting glucose, and fructosamine in Senagalese individuals with and without sickle-cell trait. PLoS One. 2019;14(2):e0212552. doi:10.1371/journal.pone.0212552

8. Byun DJ, Wolchok JD, Rosenberg LM, Girotra M. Cancer immunotherapy-immune checkpoint blockade and associated endocrinopathies. Nat Rev Endocrinol. 2017;13(4):195-207. doi:10.1038/nrendo.2016.205

9. Stamatouli AM, Quandt Z, Perdigoto AL, et al. Collateral damage: insulin-dependent diabetes induced with checkpoint inhibitors. Diabetes. 2018;67(8):1471-1480. doi:10.2337/dbi18-0002

10. Liu J, Zhou H, Zhang Y, et al. Reporting of immune checkpoint inhibitor therapy-associated diabetes, 2015-2019. Diabetes Care. 2020;43(7):e79-e80. [Published online ahead of print, 2020 May 11]. doi:10.2337/dc20-0459

11. Barroso-Sousa R, Barry WT, Garrido-Castro AC, et al. Incidence of endocrine dysfunction following the use of different immune checkpoint inhibitor regimens: a systematic review and meta-analysis. JAMA Oncol. 2018;4(2):173-182. doi:10.1001/jamaoncol.2017.3064

12. de Filette J, Andreescu CE, Cools F, Bravenboer B, Velkeniers B. A systematic review and meta-analysis of endocrine-related adverse events associated with immune checkpoint inhibitors. Horm Metab Res. 2019;51(3):145-156. doi:10.1055/a-0843-3366

13. Hughes J, Vudattu N, Sznol M, et al. Precipitation of autoimmune diabetes with anti-PD-1 immunotherapy. Diabetes Care. 2015;38(4):e55-e57. doi:10.2337/dc14-2349

14. Clotman K, Janssens K, Specenier P, Weets I, De block CEM. Programmed cell death-1 inhibitor-induced type 1 diabetes mellitus. J Clin Endocrinol Metab. 2018;103(9):3144-3154. doi:10.1210/jc.2018-00728

15. Kotwal A, Haddox C, Block M, Kudva YC. Immune checkpoint inhibitors: an emerging cause of insulin-dependent diabetes. BMJ Open Diabetes Res Care. 2019;7(1):e000591. doi:10.1136/bmjdrc-2018-000591

16. Chang LS, Barroso-Sousa R, Tolaney SM, Hodi FS, Kaiser UB, Min L. Endocrine toxicity of cancer immunotherapy targeting immune checkpoints. Endocr Rev. 2019;40(1):17-65. doi:10.1210/er.2018-00006

17. Brahmer JR, Lacchetti C, Schneider BJ, et al; National Comprehensive Cancer Network. Management of immune-related adverse events in patients treated with immune checkpoint inhibitor therapy: American Society of Clinical Oncology Clinical Practice Guideline. J Clin Oncol. 2018;36(17):1714-1768. doi:10.1200/JCO.2017.77.6385

18. Aleksova J, Lau PK, Soldatos G, Mcarthur G. Glucocorticoids did not reverse type 1 diabetes mellitus secondary to pembrolizumab in a patient with metastatic melanoma. BMJ Case Rep. 2016;2016:bcr2016217454. doi:10.1136/bcr-2016-217454

19. Afzal MZ, Mercado RR, Shirai K. Efficacy of metformin in combination with immune checkpoint inhibitors (anti-PD-1/anti-CTLA-4) in metastatic malignant melanoma. J Immunother Cancer. 2018;6(1):64. doi:10.1186/s40425-018-0375-1

20. Klabunde CN, Ambs A, Keating NL, et al. The role of primary care physicians in cancer care. J Gen Intern Med. 2009;24(9):1029-1036. doi:10.1007/s11606-009-1058-x

Article PDF
Author and Disclosure Information

Damond Ng is a Resident Physician in the Department of Medicine at David Geffen School of Medicine in Los Angeles, California. Jane Weinreb is Chief of the Division of Endocrinology at the Veterans Affairs (VA) Greater Los Angeles Healthcare System and a Clinical Professor in the Department of Medicine at University of California Los Angeles. Sara-Megumi Rumrill is an Assistant Clinical Professor in both the Division of General Internal Medicine at the San Francisco VA Medical Center and the Department of Medicine at the University of California, San Francisco.
Correspondence: Damond Ng ([email protected])

Author contributions
Damond Ng researched the data and wrote the manuscript. Sara-Megumi Rumrill and Jane Weinreb researched the data and reviewed and edited the manuscript. Damond Ng is the guarantor of this work.

Author disclosures
The authors report no actual or potential conflicts of interest with regard to this article.

Disclaimer
The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review the complete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.

Issue
Federal Practitioner - 38(11)a
Publications
Topics
Page Number
520-523
Sections
Author and Disclosure Information

Damond Ng is a Resident Physician in the Department of Medicine at David Geffen School of Medicine in Los Angeles, California. Jane Weinreb is Chief of the Division of Endocrinology at the Veterans Affairs (VA) Greater Los Angeles Healthcare System and a Clinical Professor in the Department of Medicine at University of California Los Angeles. Sara-Megumi Rumrill is an Assistant Clinical Professor in both the Division of General Internal Medicine at the San Francisco VA Medical Center and the Department of Medicine at the University of California, San Francisco.
Correspondence: Damond Ng ([email protected])

Author contributions
Damond Ng researched the data and wrote the manuscript. Sara-Megumi Rumrill and Jane Weinreb researched the data and reviewed and edited the manuscript. Damond Ng is the guarantor of this work.

Author disclosures
The authors report no actual or potential conflicts of interest with regard to this article.

Disclaimer
The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review the complete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.

Author and Disclosure Information

Damond Ng is a Resident Physician in the Department of Medicine at David Geffen School of Medicine in Los Angeles, California. Jane Weinreb is Chief of the Division of Endocrinology at the Veterans Affairs (VA) Greater Los Angeles Healthcare System and a Clinical Professor in the Department of Medicine at University of California Los Angeles. Sara-Megumi Rumrill is an Assistant Clinical Professor in both the Division of General Internal Medicine at the San Francisco VA Medical Center and the Department of Medicine at the University of California, San Francisco.
Correspondence: Damond Ng ([email protected])

Author contributions
Damond Ng researched the data and wrote the manuscript. Sara-Megumi Rumrill and Jane Weinreb researched the data and reviewed and edited the manuscript. Damond Ng is the guarantor of this work.

Author disclosures
The authors report no actual or potential conflicts of interest with regard to this article.

Disclaimer
The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review the complete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.

Article PDF
Article PDF
Low C-peptide levels should prompt a high suspicion for immune checkpoint inhibitor-induced type 1 diabetes, and initiation of insulin therapy should be strongly considered.
Low C-peptide levels should prompt a high suspicion for immune checkpoint inhibitor-induced type 1 diabetes, and initiation of insulin therapy should be strongly considered.

Immune checkpoint inhibitors (CPIs) have revolutionized cancer therapy and improved the prognosis for a variety of advanced solid tumors and Hodgkin lymphoma, but evidence is growing regarding severe endocrine disturbances.1,2 CPIs block inhibitory molecules on activated T cells to increase tumor cell destruction but also can breach normal tolerance, resulting in a spectrum of immune-related adverse events (irAE).1,2 Programmed cell death-1 (PD-1) inhibitors are one type of CPIs. Pembrolizumab is a humanized monoclonal antibody that targets the PD-1 checkpoint pathway and is approved for the treatment of malignant melanoma and non-small cell lung cancer.3,4 When the PD-1 checkpoint pathway is inhibited, T cells targeting cancer are activated, as are autoreactive T cells, such as those regulating pancreatic islet cell survival, which can lead to type 1 diabetes mellitus (T1DM).5

Case Presentation

A 95-year-old male veteran with long-standing, stable prediabetes was treated with pembrolizumab for stage 4 melanoma. Four months after treatment initiation and 3 weeks after completion of his sixth treatment cycle of pembrolizumab (2 mg/kg every 3 weeks), he presented for surveillance positron emission tomography (PET) and was incidentally found to have a serum glucose of 423 mg/dL. Hypothesis-driven history taking revealed polyuria, polydipsia, and a 12-lb weight loss during the previous 3 months. The patient reported no abdominal pain, nausea, or vomiting. He showed no evidence of pancreatic metastases on recent imaging. His family history was notable for a daughter with T1DM diagnosed at a young age.

On examination, the patient’s vital signs were normal aside from a blood pressure of 80/40 mm Hg. His body mass index was 30. He was alert and oriented with comfortable respirations and no Kussmaul breathing. He exhibited dry mucous membranes and poor skin turgor. Laboratory studies revealed 135 mmol/L sodium (reference, 135-145), 4.6 mmol/L potassium (reference, 3.6-5.2), 100 mmol/L chloride (reference, 99-106), bicarbonate of 26.5 mmol/L (reference, 23-29), serum blood urea nitrogen 27 mg/dL (reference, 6-24), 1.06 mg/dL creatinine (reference, 0.74-1.35), and 423 mg/dL glucose (reference, 70-100), with negative urine ketones. Further studies demonstrated 462 µmol/L fructosamine (reference, 190-270), correlating with hemoglobin A1c (HbA1c) close to 11.0% (HbA1c was drawn on admission but cancelled by the laboratory for unknown reasons).6,7 Later, an inappropriately low C-peptide level of 0.56 ng/mL (reference, 0.8-3.85) and a negative antiglutamic acid decarboxylase (GAD) antibody titer resulted. The patient was given IV hydration and admitted to the hospital. With input from endocrinology, the patient was started on 0.3 units per kg of body weight basal-prandial insulin therapy. Pembrolizumab was held. Six weeks after discharge, his HbA1c was 7.2%, and C-peptide improved to 1.95 ng/mL and plasma glucose 116 mg/dL. After shared decision making with his health care team, the patient decided against restarting pembrolizumab. The patient reported that his functional status was preserved, and he preferred to take fewer medications at his advanced age. He died comfortably 6 months after this presentation from complications of metastatic melanoma.

Dicussion

Immunotherapy is now an integral part of cancer treatment and can result in endocrine disturbances.1,2 Life-threatening irAEs are rare and may mimic more common conditions; thus, there is growing recognition of the need to educate health care professionals in appropriate screening and management of these conditions. CPI-induced T1DM is an uncommon but clinically significant event with an incidence of 0.4 to 1.27% and a median onset of 20 weeks after initiation of therapy (range, 1-228 weeks).8-12In case seriesfrom 3 academic centers, 59 to81% of patients with CPI-induced T1DM presented with diabetic ketoacidosis (DKA), and only 40 to 71% of patients were autoantibody positive.13-16 These patients are older than those presenting with classic T1DM, often require intensive care unit admission, and nearly invariably require exogenous insulin injections for metabolic control.13-16

Based on the later age of onset of cancers that may be treated with CPI, patients with CPI-induced T1DM may be misdiagnosed with T2DM or hyperglycemia from other causes, such as medications or acute illness in the outpatient setting, risking suboptimal treatment.

Given the infrequent incidence and lack of controlled trials, screening and treatment recommendations for CPI-induced T1DM are based on principles derived from case series and expert opinion. Development of polyuria, polydipsia, weight loss, nausea, and/or vomiting should prompt investigation for possible development or worsening of hyperglycemia, suggestive of development of T1DM.17 American Society of Clinical Oncology (ASCO) guidelines recommend that serum glucose be assessed at baseline and with each treatment cycle during induction for 12 weeks, then every 3 to 6 weeks thereafter.17 There is no reported association between the number of CPI treatments and the development of DM.8,9 Following our patient’s fifth pembrolizumab cycle, a random glucose reading was noted to be 186 mg/dL (Figure 1). Under the ASCO guidelines, ideally the patient would have received close clinical follow-up given the striking increase in plasma glucose compared with prior baseline lower values and perhaps been further evaluated with an anti-GAD antibody titer to screen for T1DM.17

Glycemic Markers During Pembrolizumab Treatmenta Figure


This patient's case adds to the published reports of CPI-induced T1DM without DKA and represents the oldest patient experiencing this irAE in the literature.13-16 The degree of elevation of his initial fructosamine, which is comparable to an average plasma glucose of approximately 270 mg/dL, belied the rapid rate of rise of his recent plasma glucose. Given the trajectory of glycemic markers and symptoms, one could certainly be concerned about imminent decompensation to DKA. However, fortuitous point-of-care glucose reading prior to surveillance PET resulted in a new critical diagnosis and initiation of treatment.

 

 



Assessing the need for inpatient evaluation includes obtaining urine ketones and acid-base status as screening for DKA.17 Antibodies and C-peptide can be sent to support diagnosis of new onset T1DM, although the initiation of therapy should not be delayed for these results.17 As noted before, many of these patients also are antibody negative.13-16 Low C-peptide levels should prompt a high suspicion for CPI-induced T1DM, and initiation of insulin therapy should be strongly considered.17 In a case series of 27 patients, 85% exhibited a rapid loss of β-cell function, evidenced by the acute progression to hyperglycemia and low or undetectable levels of C-peptide at diagnosis.9 Likewise, our patient had a low C-peptide level and negative anti-GAD antibody titer but was treated before these results were available. Inpatient admission for close glycemic monitoring may be reasonable; several cases reported prompt diagnosis and avoidance of DKA in this setting.17

In contrast to other irAEs, there is no available evidence that high-dose corticosteroids alter the course of pembrolizumab-induced T2DM.18 Depending on the degree of hyperglycemia, endocrinology consultation and insulin treatment are appropriate where the diagnosis of T1DM is suspected even without evidence of DKA.17 For patients with T2DM, there may be a positive synergistic effect of metformin in combination with CPIs in tumor control.19 Our patient’s C-peptide improved with insulin treatment, consistent with correction of glucose toxicity and a honeymoon period in his course. However, in patients reported with pembrolizumab-induced T1DM, insulin requirement for treatment generally persists despite cessation of pembrolizumab therapy.13-16

Conclusions

Pembrolizumab-induced T1DM is a rare, but potentially life-threatening irAE. The acute risk of DKA requires early recognition and prompt treatment of patients taking CPIs. More than 90% of primary care physicians (PCPs) fulfill general medical care roles for patients with cancer; therefore, they play an essential role in evaluating symptoms during therapy.20 Further studies evaluating the role of PCPs and outcomes when PCPs are involved in oncologic care should be conducted.

Figure of Letter

With increased index of suspicion, this clinical scenario presents an opportunity for PCPs that may help reduce irAE-associated morbidity and mortality of patients on CPIs, like pembrolizumab. Figure 2 illustrates an example addendum that can be used to alert and tag a PCP of a mutual patient after initiation of CPI therapy. Determining the optimal interface between PCPs, oncologists, and endocrinologists in delivering and coordinating high-quality cancer care in the setting of immunotherapy is an important area for ongoing quality improvement.

Acknowledgment

The authors thank all the staff and health care professionals at VA Greater Los Angeles Healthcare System who were involved in the care of this patient.

Immune checkpoint inhibitors (CPIs) have revolutionized cancer therapy and improved the prognosis for a variety of advanced solid tumors and Hodgkin lymphoma, but evidence is growing regarding severe endocrine disturbances.1,2 CPIs block inhibitory molecules on activated T cells to increase tumor cell destruction but also can breach normal tolerance, resulting in a spectrum of immune-related adverse events (irAE).1,2 Programmed cell death-1 (PD-1) inhibitors are one type of CPIs. Pembrolizumab is a humanized monoclonal antibody that targets the PD-1 checkpoint pathway and is approved for the treatment of malignant melanoma and non-small cell lung cancer.3,4 When the PD-1 checkpoint pathway is inhibited, T cells targeting cancer are activated, as are autoreactive T cells, such as those regulating pancreatic islet cell survival, which can lead to type 1 diabetes mellitus (T1DM).5

Case Presentation

A 95-year-old male veteran with long-standing, stable prediabetes was treated with pembrolizumab for stage 4 melanoma. Four months after treatment initiation and 3 weeks after completion of his sixth treatment cycle of pembrolizumab (2 mg/kg every 3 weeks), he presented for surveillance positron emission tomography (PET) and was incidentally found to have a serum glucose of 423 mg/dL. Hypothesis-driven history taking revealed polyuria, polydipsia, and a 12-lb weight loss during the previous 3 months. The patient reported no abdominal pain, nausea, or vomiting. He showed no evidence of pancreatic metastases on recent imaging. His family history was notable for a daughter with T1DM diagnosed at a young age.

On examination, the patient’s vital signs were normal aside from a blood pressure of 80/40 mm Hg. His body mass index was 30. He was alert and oriented with comfortable respirations and no Kussmaul breathing. He exhibited dry mucous membranes and poor skin turgor. Laboratory studies revealed 135 mmol/L sodium (reference, 135-145), 4.6 mmol/L potassium (reference, 3.6-5.2), 100 mmol/L chloride (reference, 99-106), bicarbonate of 26.5 mmol/L (reference, 23-29), serum blood urea nitrogen 27 mg/dL (reference, 6-24), 1.06 mg/dL creatinine (reference, 0.74-1.35), and 423 mg/dL glucose (reference, 70-100), with negative urine ketones. Further studies demonstrated 462 µmol/L fructosamine (reference, 190-270), correlating with hemoglobin A1c (HbA1c) close to 11.0% (HbA1c was drawn on admission but cancelled by the laboratory for unknown reasons).6,7 Later, an inappropriately low C-peptide level of 0.56 ng/mL (reference, 0.8-3.85) and a negative antiglutamic acid decarboxylase (GAD) antibody titer resulted. The patient was given IV hydration and admitted to the hospital. With input from endocrinology, the patient was started on 0.3 units per kg of body weight basal-prandial insulin therapy. Pembrolizumab was held. Six weeks after discharge, his HbA1c was 7.2%, and C-peptide improved to 1.95 ng/mL and plasma glucose 116 mg/dL. After shared decision making with his health care team, the patient decided against restarting pembrolizumab. The patient reported that his functional status was preserved, and he preferred to take fewer medications at his advanced age. He died comfortably 6 months after this presentation from complications of metastatic melanoma.

Dicussion

Immunotherapy is now an integral part of cancer treatment and can result in endocrine disturbances.1,2 Life-threatening irAEs are rare and may mimic more common conditions; thus, there is growing recognition of the need to educate health care professionals in appropriate screening and management of these conditions. CPI-induced T1DM is an uncommon but clinically significant event with an incidence of 0.4 to 1.27% and a median onset of 20 weeks after initiation of therapy (range, 1-228 weeks).8-12In case seriesfrom 3 academic centers, 59 to81% of patients with CPI-induced T1DM presented with diabetic ketoacidosis (DKA), and only 40 to 71% of patients were autoantibody positive.13-16 These patients are older than those presenting with classic T1DM, often require intensive care unit admission, and nearly invariably require exogenous insulin injections for metabolic control.13-16

Based on the later age of onset of cancers that may be treated with CPI, patients with CPI-induced T1DM may be misdiagnosed with T2DM or hyperglycemia from other causes, such as medications or acute illness in the outpatient setting, risking suboptimal treatment.

Given the infrequent incidence and lack of controlled trials, screening and treatment recommendations for CPI-induced T1DM are based on principles derived from case series and expert opinion. Development of polyuria, polydipsia, weight loss, nausea, and/or vomiting should prompt investigation for possible development or worsening of hyperglycemia, suggestive of development of T1DM.17 American Society of Clinical Oncology (ASCO) guidelines recommend that serum glucose be assessed at baseline and with each treatment cycle during induction for 12 weeks, then every 3 to 6 weeks thereafter.17 There is no reported association between the number of CPI treatments and the development of DM.8,9 Following our patient’s fifth pembrolizumab cycle, a random glucose reading was noted to be 186 mg/dL (Figure 1). Under the ASCO guidelines, ideally the patient would have received close clinical follow-up given the striking increase in plasma glucose compared with prior baseline lower values and perhaps been further evaluated with an anti-GAD antibody titer to screen for T1DM.17

Glycemic Markers During Pembrolizumab Treatmenta Figure


This patient's case adds to the published reports of CPI-induced T1DM without DKA and represents the oldest patient experiencing this irAE in the literature.13-16 The degree of elevation of his initial fructosamine, which is comparable to an average plasma glucose of approximately 270 mg/dL, belied the rapid rate of rise of his recent plasma glucose. Given the trajectory of glycemic markers and symptoms, one could certainly be concerned about imminent decompensation to DKA. However, fortuitous point-of-care glucose reading prior to surveillance PET resulted in a new critical diagnosis and initiation of treatment.

 

 



Assessing the need for inpatient evaluation includes obtaining urine ketones and acid-base status as screening for DKA.17 Antibodies and C-peptide can be sent to support diagnosis of new onset T1DM, although the initiation of therapy should not be delayed for these results.17 As noted before, many of these patients also are antibody negative.13-16 Low C-peptide levels should prompt a high suspicion for CPI-induced T1DM, and initiation of insulin therapy should be strongly considered.17 In a case series of 27 patients, 85% exhibited a rapid loss of β-cell function, evidenced by the acute progression to hyperglycemia and low or undetectable levels of C-peptide at diagnosis.9 Likewise, our patient had a low C-peptide level and negative anti-GAD antibody titer but was treated before these results were available. Inpatient admission for close glycemic monitoring may be reasonable; several cases reported prompt diagnosis and avoidance of DKA in this setting.17

In contrast to other irAEs, there is no available evidence that high-dose corticosteroids alter the course of pembrolizumab-induced T2DM.18 Depending on the degree of hyperglycemia, endocrinology consultation and insulin treatment are appropriate where the diagnosis of T1DM is suspected even without evidence of DKA.17 For patients with T2DM, there may be a positive synergistic effect of metformin in combination with CPIs in tumor control.19 Our patient’s C-peptide improved with insulin treatment, consistent with correction of glucose toxicity and a honeymoon period in his course. However, in patients reported with pembrolizumab-induced T1DM, insulin requirement for treatment generally persists despite cessation of pembrolizumab therapy.13-16

Conclusions

Pembrolizumab-induced T1DM is a rare, but potentially life-threatening irAE. The acute risk of DKA requires early recognition and prompt treatment of patients taking CPIs. More than 90% of primary care physicians (PCPs) fulfill general medical care roles for patients with cancer; therefore, they play an essential role in evaluating symptoms during therapy.20 Further studies evaluating the role of PCPs and outcomes when PCPs are involved in oncologic care should be conducted.

Figure of Letter

With increased index of suspicion, this clinical scenario presents an opportunity for PCPs that may help reduce irAE-associated morbidity and mortality of patients on CPIs, like pembrolizumab. Figure 2 illustrates an example addendum that can be used to alert and tag a PCP of a mutual patient after initiation of CPI therapy. Determining the optimal interface between PCPs, oncologists, and endocrinologists in delivering and coordinating high-quality cancer care in the setting of immunotherapy is an important area for ongoing quality improvement.

Acknowledgment

The authors thank all the staff and health care professionals at VA Greater Los Angeles Healthcare System who were involved in the care of this patient.

References

1. Puzanov I, Diab A, Abdallah K, et al; Society for Immunotherapy of Cancer Toxicity Management Working Group. Managing toxicities associated with immune checkpoint inhibitors: consensus recommendations from the Society for Immunotherapy of Cancer (SITC) Toxicity Management Working Group. J Immunother Cancer. 2017;5(1):95. doi:10.1186/s40425-017-0300-z

2. Villa NM, Farahmand A, Du L, et al. Endocrinopathies with use of cancer immunotherapies. Clin Endocrinol (Oxf). 2018;88(2):327-332. doi:10.1111/cen.13483

3. Schachter J, Ribas A, Long GV, et al. Pembrolizumab versus ipilimumab for advanced melanoma: final overall survival results of a multicentre, randomised, open-label phase 3 study (KEYNOTE-006). Lancet. 2017;390(10105):1853-1862. doi:10.1016/S0140-6736(17)31601-X

4. Garon EB, Hellmann MD, Rizvi NA, et al. Five-year overall survival for patients with advanced non-small-cell lung cancer treated with pembrolizumab: results from the phase I KEYNOTE-001 Study. J Clin Oncol. 2019;37(28):2518-2527. doi:10.1200/JCO.19.00934

5. Ribas A. Tumor immunotherapy directed at PD-1. N Engl J Med. 2012;366(26):2517-2519. doi:10.1056/NEJMe1205943

6. Malmstrom H, Walldius G, Grill V, Jungner I, Gudbjomsdottir S, Hammar N. Frustosamine is a useful indicator of hyperglycemia and glucose control in clinical and epidemiological studies- cross-sectional and longitudinal experience from the AMORIS cohort. PLoS One. 2014;9(10):e111463. doi:10.1371/journal.pone.0111463

7. Skinner S, Diaw M, Mbaye MN, et al. Evaluation of agreement between hemoglobin A1c, fasting glucose, and fructosamine in Senagalese individuals with and without sickle-cell trait. PLoS One. 2019;14(2):e0212552. doi:10.1371/journal.pone.0212552

8. Byun DJ, Wolchok JD, Rosenberg LM, Girotra M. Cancer immunotherapy-immune checkpoint blockade and associated endocrinopathies. Nat Rev Endocrinol. 2017;13(4):195-207. doi:10.1038/nrendo.2016.205

9. Stamatouli AM, Quandt Z, Perdigoto AL, et al. Collateral damage: insulin-dependent diabetes induced with checkpoint inhibitors. Diabetes. 2018;67(8):1471-1480. doi:10.2337/dbi18-0002

10. Liu J, Zhou H, Zhang Y, et al. Reporting of immune checkpoint inhibitor therapy-associated diabetes, 2015-2019. Diabetes Care. 2020;43(7):e79-e80. [Published online ahead of print, 2020 May 11]. doi:10.2337/dc20-0459

11. Barroso-Sousa R, Barry WT, Garrido-Castro AC, et al. Incidence of endocrine dysfunction following the use of different immune checkpoint inhibitor regimens: a systematic review and meta-analysis. JAMA Oncol. 2018;4(2):173-182. doi:10.1001/jamaoncol.2017.3064

12. de Filette J, Andreescu CE, Cools F, Bravenboer B, Velkeniers B. A systematic review and meta-analysis of endocrine-related adverse events associated with immune checkpoint inhibitors. Horm Metab Res. 2019;51(3):145-156. doi:10.1055/a-0843-3366

13. Hughes J, Vudattu N, Sznol M, et al. Precipitation of autoimmune diabetes with anti-PD-1 immunotherapy. Diabetes Care. 2015;38(4):e55-e57. doi:10.2337/dc14-2349

14. Clotman K, Janssens K, Specenier P, Weets I, De block CEM. Programmed cell death-1 inhibitor-induced type 1 diabetes mellitus. J Clin Endocrinol Metab. 2018;103(9):3144-3154. doi:10.1210/jc.2018-00728

15. Kotwal A, Haddox C, Block M, Kudva YC. Immune checkpoint inhibitors: an emerging cause of insulin-dependent diabetes. BMJ Open Diabetes Res Care. 2019;7(1):e000591. doi:10.1136/bmjdrc-2018-000591

16. Chang LS, Barroso-Sousa R, Tolaney SM, Hodi FS, Kaiser UB, Min L. Endocrine toxicity of cancer immunotherapy targeting immune checkpoints. Endocr Rev. 2019;40(1):17-65. doi:10.1210/er.2018-00006

17. Brahmer JR, Lacchetti C, Schneider BJ, et al; National Comprehensive Cancer Network. Management of immune-related adverse events in patients treated with immune checkpoint inhibitor therapy: American Society of Clinical Oncology Clinical Practice Guideline. J Clin Oncol. 2018;36(17):1714-1768. doi:10.1200/JCO.2017.77.6385

18. Aleksova J, Lau PK, Soldatos G, Mcarthur G. Glucocorticoids did not reverse type 1 diabetes mellitus secondary to pembrolizumab in a patient with metastatic melanoma. BMJ Case Rep. 2016;2016:bcr2016217454. doi:10.1136/bcr-2016-217454

19. Afzal MZ, Mercado RR, Shirai K. Efficacy of metformin in combination with immune checkpoint inhibitors (anti-PD-1/anti-CTLA-4) in metastatic malignant melanoma. J Immunother Cancer. 2018;6(1):64. doi:10.1186/s40425-018-0375-1

20. Klabunde CN, Ambs A, Keating NL, et al. The role of primary care physicians in cancer care. J Gen Intern Med. 2009;24(9):1029-1036. doi:10.1007/s11606-009-1058-x

References

1. Puzanov I, Diab A, Abdallah K, et al; Society for Immunotherapy of Cancer Toxicity Management Working Group. Managing toxicities associated with immune checkpoint inhibitors: consensus recommendations from the Society for Immunotherapy of Cancer (SITC) Toxicity Management Working Group. J Immunother Cancer. 2017;5(1):95. doi:10.1186/s40425-017-0300-z

2. Villa NM, Farahmand A, Du L, et al. Endocrinopathies with use of cancer immunotherapies. Clin Endocrinol (Oxf). 2018;88(2):327-332. doi:10.1111/cen.13483

3. Schachter J, Ribas A, Long GV, et al. Pembrolizumab versus ipilimumab for advanced melanoma: final overall survival results of a multicentre, randomised, open-label phase 3 study (KEYNOTE-006). Lancet. 2017;390(10105):1853-1862. doi:10.1016/S0140-6736(17)31601-X

4. Garon EB, Hellmann MD, Rizvi NA, et al. Five-year overall survival for patients with advanced non-small-cell lung cancer treated with pembrolizumab: results from the phase I KEYNOTE-001 Study. J Clin Oncol. 2019;37(28):2518-2527. doi:10.1200/JCO.19.00934

5. Ribas A. Tumor immunotherapy directed at PD-1. N Engl J Med. 2012;366(26):2517-2519. doi:10.1056/NEJMe1205943

6. Malmstrom H, Walldius G, Grill V, Jungner I, Gudbjomsdottir S, Hammar N. Frustosamine is a useful indicator of hyperglycemia and glucose control in clinical and epidemiological studies- cross-sectional and longitudinal experience from the AMORIS cohort. PLoS One. 2014;9(10):e111463. doi:10.1371/journal.pone.0111463

7. Skinner S, Diaw M, Mbaye MN, et al. Evaluation of agreement between hemoglobin A1c, fasting glucose, and fructosamine in Senagalese individuals with and without sickle-cell trait. PLoS One. 2019;14(2):e0212552. doi:10.1371/journal.pone.0212552

8. Byun DJ, Wolchok JD, Rosenberg LM, Girotra M. Cancer immunotherapy-immune checkpoint blockade and associated endocrinopathies. Nat Rev Endocrinol. 2017;13(4):195-207. doi:10.1038/nrendo.2016.205

9. Stamatouli AM, Quandt Z, Perdigoto AL, et al. Collateral damage: insulin-dependent diabetes induced with checkpoint inhibitors. Diabetes. 2018;67(8):1471-1480. doi:10.2337/dbi18-0002

10. Liu J, Zhou H, Zhang Y, et al. Reporting of immune checkpoint inhibitor therapy-associated diabetes, 2015-2019. Diabetes Care. 2020;43(7):e79-e80. [Published online ahead of print, 2020 May 11]. doi:10.2337/dc20-0459

11. Barroso-Sousa R, Barry WT, Garrido-Castro AC, et al. Incidence of endocrine dysfunction following the use of different immune checkpoint inhibitor regimens: a systematic review and meta-analysis. JAMA Oncol. 2018;4(2):173-182. doi:10.1001/jamaoncol.2017.3064

12. de Filette J, Andreescu CE, Cools F, Bravenboer B, Velkeniers B. A systematic review and meta-analysis of endocrine-related adverse events associated with immune checkpoint inhibitors. Horm Metab Res. 2019;51(3):145-156. doi:10.1055/a-0843-3366

13. Hughes J, Vudattu N, Sznol M, et al. Precipitation of autoimmune diabetes with anti-PD-1 immunotherapy. Diabetes Care. 2015;38(4):e55-e57. doi:10.2337/dc14-2349

14. Clotman K, Janssens K, Specenier P, Weets I, De block CEM. Programmed cell death-1 inhibitor-induced type 1 diabetes mellitus. J Clin Endocrinol Metab. 2018;103(9):3144-3154. doi:10.1210/jc.2018-00728

15. Kotwal A, Haddox C, Block M, Kudva YC. Immune checkpoint inhibitors: an emerging cause of insulin-dependent diabetes. BMJ Open Diabetes Res Care. 2019;7(1):e000591. doi:10.1136/bmjdrc-2018-000591

16. Chang LS, Barroso-Sousa R, Tolaney SM, Hodi FS, Kaiser UB, Min L. Endocrine toxicity of cancer immunotherapy targeting immune checkpoints. Endocr Rev. 2019;40(1):17-65. doi:10.1210/er.2018-00006

17. Brahmer JR, Lacchetti C, Schneider BJ, et al; National Comprehensive Cancer Network. Management of immune-related adverse events in patients treated with immune checkpoint inhibitor therapy: American Society of Clinical Oncology Clinical Practice Guideline. J Clin Oncol. 2018;36(17):1714-1768. doi:10.1200/JCO.2017.77.6385

18. Aleksova J, Lau PK, Soldatos G, Mcarthur G. Glucocorticoids did not reverse type 1 diabetes mellitus secondary to pembrolizumab in a patient with metastatic melanoma. BMJ Case Rep. 2016;2016:bcr2016217454. doi:10.1136/bcr-2016-217454

19. Afzal MZ, Mercado RR, Shirai K. Efficacy of metformin in combination with immune checkpoint inhibitors (anti-PD-1/anti-CTLA-4) in metastatic malignant melanoma. J Immunother Cancer. 2018;6(1):64. doi:10.1186/s40425-018-0375-1

20. Klabunde CN, Ambs A, Keating NL, et al. The role of primary care physicians in cancer care. J Gen Intern Med. 2009;24(9):1029-1036. doi:10.1007/s11606-009-1058-x

Issue
Federal Practitioner - 38(11)a
Issue
Federal Practitioner - 38(11)a
Page Number
520-523
Page Number
520-523
Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Annular Erythema of Infancy With Reactive Helper T Lymphocytes

Article Type
Changed
Sun, 11/07/2021 - 15:23
Display Headline
Annular Erythema of Infancy With Reactive Helper T Lymphocytes

Annular erythemas of infancy (AEIs) are rare benign skin eruptions characterized by annular or circinate, erythematous patches and plaques that arise in patients younger than 1 year.1 Annular erythemas of infancy originally were described by Peterson and Jarratt2 in 1981. Relatively few cases of AEIs have been reported in the literature (eTable).2-15

 

Case Report

An 11-month-old girl presented to dermatology for a rash characterized by annular erythematous patches and plaques on the back, arms, and legs (Figure 1). Three months prior, the rash was more diffuse, monomorphic, and papular. Based on physical examination, the differential diagnosis included a gyrate erythema such as erythema annulare centrifugum (EAC), neonatal lupus, a viral exanthem, leukemia cutis, and AEI. A skin punch biopsy was performed.

FIGURE 1. A, An 11-month-old girl with annular erythematous patches and plaques on the back. B, Annular erythematous lesions were present on the right arm, from which a punch biopsy was taken.

Histologically, the biopsy revealed a superficial to mid dermal, tight, coat sleeve–like, perivascular lymphohistiocytic infiltrate admixed with rare neutrophils in eosinophils within the dermis (Figure 2A). The infiltrate also contained numerous large mononuclear cells with enlarged nuclei, fine loose chromatin, rare nucleoli, and a thin rim of cytoplasm (Figure 2B). There were associated apoptotic bodies with karyorrhectic debris. Immunohistochemistry exhibited enlarged cells that were strong staining with CD3 and CD4, which was consistent with reactive helper T cells (Figure 3). A myeloperoxidase stain highlighted few neutrophils. Stains for terminal deoxynucleotidyl transferase, CD1a, CD117, and CD34 were negative. These findings along with the clinical presentation yielded a diagnosis of AEI with reactive helper T cells.

FIGURE 2. Histopathology demonstrated annular erythema of infancy with mononuclear cells. A, There was a superficial to mid dermal, tight, coat sleeve–like, perivascular, lymphohistiocytic infiltrate admixed with rare neutrophils in eosinophils within the dermis (H&E, original magnification ×40). B, The infiltrate contained numerous large mononuclear cells with enlarged nuclei, fine loose chromatin, rare nucleoli, and a thin rim of cytoplasm (H&E, original magnification ×400).

Comment

Clinical Presentation of AEIs—Annular erythemas of infancy are rare benign skin eruptions that develop in the first few months of life.1,16 Few cases have been reported (eTable). Clinically, AEIs are characterized by annular or circinate, erythematous patches and plaques. They can occur on the face, trunk, and extremities, and they completely resolve by 1 year of age in most cases. One case was reported to persist in a patient from birth until 15 years of age.9 It is thought that AEIs may occur as a hypersensitivity reaction to an unrecognized antigen.

FIGURE 3. A, Immunohistochemistry revealed the infiltrate was composed predominantly of CD3+ T lymphocytes (original magnification ×100). B, The enlarged cells were CD4+, consistent with reactive helper T cells (original magnification ×400).

Histopathology—Histologically, AEIs demonstrate a superficial and deep, perivascular, inflammatory infiltrate in the dermis composed of small lymphocytes, some neutrophils, and eosinophils.16 Less common variants of AEI include eosinophilic annular erythema, characterized by a diffuse dermal infiltrate of eosinophils and some lymphocytes, and neutrophilic figurate erythema of infancy, characterized by a dermal infiltrate with neutrophils and leukocytoclasis without vasculitis.1

Our patient’s skin rash was unusual in that the biopsy demonstrated few neutrophils, rare eosinophils, and larger mononuclear cells consistent with reactive helper T lymphocytes. Although these cells may raise concern for an atypical lymphoid infiltrate, recognition of areas with more conventional histopathology of AEIs can facilitate the correct diagnosis.

Differential Diagnosis—The main considerations in the differential diagnosis for AEIs include the following: EAC, familial annular erythema, erythema gyratum atrophicans transiens neonatale, erythema chronicum migrans, urticaria, tinea corporis, neonatal lupus erythematosus, viral exanthems, and leukemia cutis.16

 

 

Erythema annulare centrifugum typically begins in middle age and follows a course of 2 or more years.2 It occurs in association with an underlying infection or neoplasm, and it can develop on the trunk and proximal extremities. Morphologically, EAC can present with arcuate or polycyclic lesions with trailing scale. Histologically, a skin biopsy shows a tight, coat sleeve–like, perivascular, lymphohistiocytic infiltrate in the dermis, with variable epidermal spongiosis and parakeratosis.16 Our patient’s biopsy did show a tight perivascular infiltrate, raising suspicion for EAC. However, the eruption occurred in infancy, and she had no clinical evidence of infection or neoplasm.

Familial annular erythemas can arise within a few days after birth and can present on any part of the body, including the tongue.2 Individual lesions can persist for 4 to 5 days and can accompany congenital malformations. Morphologically, they can present as papules that slowly enlarge to form arcuate lesions with central hyperpigmentation. Histologically, there can be a mild, perivascular, lymphocytic infiltrate in the dermis.16 Our patient’s lesions showed no scale or pigmentation and occurred without a family history or associated malformations.

Erythema gyratum atrophicans transiens neonatale also can arise in the first few days of life and can affect the trunk, neck, and lips.16 Morphologically, the skin lesions can present as arcuate erythematous patches (3–20 mm) with raised borders and central atrophy. Histologically, there is epidermal atrophy with a dermal perivascular mononuclear cell infiltrate with edema. Our patient’s clinical presentation was not classic for this condition, and the lesions showed no atrophy.

Erythema chronicum migrans can arise in children, often with a history of an arthropod bite.13 Morphologically, lesions can evolve over weeks to months and rarely are multiple. Erythema chronicum migrans most commonly occurs in the United States in association with Lyme disease from infection with Borrelia burgdorferi. Histologically, erythema chronicum migrans shows a superficial and deep, perivascular lymphocytic infiltrate in the dermis with plasma cells and eosinophils. A silver stain can demonstrate dermal spirochetes. Our patient had no history of an arthropod bite. A Warthin-Starry stain performed on the biopsy was negative for spirochetes, and serologies for Lyme disease were negative.

Urticaria is rare in neonates and can occur on any part of the body.2 Morphologically, the skin lesions can present as arcuate, erythematous, and polycyclic plaques that wax and wane. Histologically, there is dermal edema with a mild, perivascular and interstitial, mixed inflammatory infiltrate.16 Our patient’s biopsy did not reveal notable edema, and the perivascular infiltrate was coat sleeve–like with few neutrophils and eosinophils. The patient did not respond to initial treatment with antihistamines, making urticaria less likely.

 

 

Tinea corporis is rare in neonates and can occur on any part of the body.13 Morphologically, it can present as scaly annular lesions that are fixed and more persistent. Histologically, there are fungal hyphae and/or yeast in the stratum corneum with spongiotic dermatitis and parakeratosis. Our patient’s lesions were not scaly, and the biopsy demonstrated minimal spongiosis. A periodic acid–Schiff special stain was negative for fungal microorganisms.

Neonatal lupus erythematosus can arise at birth or during the first few weeks of life.16 Morphologically, the skin lesions occur on the scalp, forehead, or neck in a periorbital or malar distribution. They can present as erythematous, annular, scaly patches and plaques. Transplacental transmission of material autoantibodies has been implicated in the etiology, and a complication is infantile heart block. Histologically, a skin biopsy typically shows interface/lichenoid dermatitis. However, our patient’s biopsy did not demonstrate interface changes, and serologically she was negative for autoantibodies.

Viral exanthems are skin eruptions that accompany underlying viral infections.17 Morphologically, patients can present with an erythematous maculopapular rash, sometimes with vesicular, petechial, and urticarial lesions. Laboratory confirmation is made by virus-specific serologies. Histologically, viral exanthems can show a superficial, perivascular, lymphocytic infiltrate in the dermis, with reactive T cells and epidermal spongiosis. Our patient was afebrile and had no known sick contacts. A cytomegalovirus immunohistochemical study on the biopsy was negative, and an Epstein-Barr encoding region in situ hybridization study was negative.

Leukemia cutis is the infiltration of the skin by leukemic cells, most often in conjunction with systemic leukemia.18 In infants and children, the most common leukemia is B-cell acute lymphoblastic leukemia. Morphologically, the skin lesions are characterized by single or multiple violaceous papules, nodules, and plaques. Histologically, there is a perivascular to interstitial infiltrate of atypical mononuclear cells in the dermis and sometimes subcutis. The leukemic cells demonstrate enlarged nuclei with coarse chromatin and prominent nucleoli. Increased mitotic activity may be seen with karyorrhectic debris. Immunohistochemically, the tumor cells can be positive for myeloperoxidase, CD43, CD68, CD34, and CD117.18 Although our patient’s biopsy demonstrated mononuclear cells with karyorrhexis, the cells did not have striking atypia and were negative for blast markers. A recent complete blood cell count on the patient was normal.

Conclusion

We report an unusual case of AEI with mononuclear cells consistent with helper T cells. One must keep these cells in mind when evaluating a biopsy of AEI, as they are benign and not suggestive of an atypical lymphoid infiltrate or leukemia cutis. This will prevent misdiagnosis and ensure that the patient receives appropriate management.

References
  1. Ríos-Martín JJ, Ferrándiz-Pulido L, Moreno-Ramírez D. Approaches to the dermatopathologic diagnosis of figurate lesions [in Spanish]. Actas Dermosifiliogr. 2011;102:316-324. doi:10.1016/j.ad.2010.12.009
  2. Peterson AO, Jarratt M. Annular erythema of infancy. Arch Dermatol. 1981;117:145-148.
  3. Toonstra J, de Wit RF. “Persistent” annular erythema of infancy. Arch Dermatol.1984;120:1069-1072.
  4. Hebert AA, Esterly NB. Annular erythema of infancy. J Am Acad Dermatol. 1986;14:339-343.
  5. Cox NH, McQueen A, Evans TJ, et al. An annular erythema of infancy. Arch Dermatol. 1987;123:510-513.
  6. Helm TN, Bass J, Chang LW, et al. Persistent annular erythema of infancy. Pediatr Dermatol. 1993;10:46-48.
  7. Kunz M, Hamm K, Bröcker EB, et al. Annular erythema in childhood—a new eosinophilic dermatosis [in German]. Hautarzt. 1998;49:131-134.
  8. Stachowitz S, Abeck D, Schmidt T, et al. Persistent annular erythema of infancy associated with intestinal Candida colonization. Clin Exp Dermatol. 2000;25:404-405.
  9. Wong L-C, Kakakios A, Rogers M. Congenital annular erythema persisting in a 15-year-old girl. Australas J Dermatol. 2002;43:55-61.
  10. Patrizi A, Savoia F, Varotti E, et al. Neutrophilic figurate erythema of infancy. Pediatr Dermatol. 2008;25:255-260. doi:10.1111/j.1525-1470.2008.00646.x
  11. Saha A, Seth J, Mukherjee S, et al. Annular erythema of infancy: a diagnostic challenge. Indian J Paediatr Dermatol. 2014;15:147-149. doi:10.4103/2319-7250.143678
  12. Pfingstler LF, Miller KP, Pride H. Recurring diffuse annular erythematous plaques in a newborn. JAMA Dermatol. 2014;150:565-566. doi:10.1001/jamadermatol.2013.8059
  13. Del Puerto Troncoso C, Curi Tuma M, González Bombardiere S, et al. Neutrophilic figurate erythema of infancy associated with juvenile myelomonocytic leukemia. Actas Dermosifiliogr. 2015;106:431-433. doi:10.1016/j.ad.2014.09.013
  14. Hamidi S, Prose NS, Selim MA. Neutrophilic figurate erythema of infancy: a diagnostic challenge [published online December 26, 2018]. J Cutan Pathol. 2019;46:216-220. doi:10.1111/cup.13394
  15. Patel N, Goldbach H, Hogeling M. An annular eruption in a young child. JAMA Dermatol. 2018;154:1213-1214. doi:10.1001/jamadermatol.2018.1174
  16. Palit A, Inamadar AC. Annular, erythematous skin lesions in a neonate. Indian Dermatol Online J. 2012;3:45-47. doi:10.4103/2229-5178.93504
  17. Keighley CL, Saunderson RB, Kok J, et al. Viral exanthems. Curr Opin Infect Dis. 2015;28:139-150. doi:10.1097/QCO.0000000000000145
  18. Cronin DMP, George TI, Sundram UN. An updated approach to the diagnosis of myeloid leukemia cutis. Am J Clin Pathol. 2009;132:101-110. doi:10.1309/AJCP6GR8BDEXPKHR
Article PDF
Author and Disclosure Information

From the University of California, Los Angeles, David Geffen School of Medicine.

The authors report no conflict of interest.

The eTable is available in the Appendix online at www.mdedge.com/dermatology.

Correspondence: Patrick Tran, MD, 1000 W Carson St, Torrance, CA 90502 ([email protected]).

Issue
Cutis - 108(5)
Publications
Topics
Page Number
289-E2
Sections
Author and Disclosure Information

From the University of California, Los Angeles, David Geffen School of Medicine.

The authors report no conflict of interest.

The eTable is available in the Appendix online at www.mdedge.com/dermatology.

Correspondence: Patrick Tran, MD, 1000 W Carson St, Torrance, CA 90502 ([email protected]).

Author and Disclosure Information

From the University of California, Los Angeles, David Geffen School of Medicine.

The authors report no conflict of interest.

The eTable is available in the Appendix online at www.mdedge.com/dermatology.

Correspondence: Patrick Tran, MD, 1000 W Carson St, Torrance, CA 90502 ([email protected]).

Article PDF
Article PDF

Annular erythemas of infancy (AEIs) are rare benign skin eruptions characterized by annular or circinate, erythematous patches and plaques that arise in patients younger than 1 year.1 Annular erythemas of infancy originally were described by Peterson and Jarratt2 in 1981. Relatively few cases of AEIs have been reported in the literature (eTable).2-15

 

Case Report

An 11-month-old girl presented to dermatology for a rash characterized by annular erythematous patches and plaques on the back, arms, and legs (Figure 1). Three months prior, the rash was more diffuse, monomorphic, and papular. Based on physical examination, the differential diagnosis included a gyrate erythema such as erythema annulare centrifugum (EAC), neonatal lupus, a viral exanthem, leukemia cutis, and AEI. A skin punch biopsy was performed.

FIGURE 1. A, An 11-month-old girl with annular erythematous patches and plaques on the back. B, Annular erythematous lesions were present on the right arm, from which a punch biopsy was taken.

Histologically, the biopsy revealed a superficial to mid dermal, tight, coat sleeve–like, perivascular lymphohistiocytic infiltrate admixed with rare neutrophils in eosinophils within the dermis (Figure 2A). The infiltrate also contained numerous large mononuclear cells with enlarged nuclei, fine loose chromatin, rare nucleoli, and a thin rim of cytoplasm (Figure 2B). There were associated apoptotic bodies with karyorrhectic debris. Immunohistochemistry exhibited enlarged cells that were strong staining with CD3 and CD4, which was consistent with reactive helper T cells (Figure 3). A myeloperoxidase stain highlighted few neutrophils. Stains for terminal deoxynucleotidyl transferase, CD1a, CD117, and CD34 were negative. These findings along with the clinical presentation yielded a diagnosis of AEI with reactive helper T cells.

FIGURE 2. Histopathology demonstrated annular erythema of infancy with mononuclear cells. A, There was a superficial to mid dermal, tight, coat sleeve–like, perivascular, lymphohistiocytic infiltrate admixed with rare neutrophils in eosinophils within the dermis (H&E, original magnification ×40). B, The infiltrate contained numerous large mononuclear cells with enlarged nuclei, fine loose chromatin, rare nucleoli, and a thin rim of cytoplasm (H&E, original magnification ×400).

Comment

Clinical Presentation of AEIs—Annular erythemas of infancy are rare benign skin eruptions that develop in the first few months of life.1,16 Few cases have been reported (eTable). Clinically, AEIs are characterized by annular or circinate, erythematous patches and plaques. They can occur on the face, trunk, and extremities, and they completely resolve by 1 year of age in most cases. One case was reported to persist in a patient from birth until 15 years of age.9 It is thought that AEIs may occur as a hypersensitivity reaction to an unrecognized antigen.

FIGURE 3. A, Immunohistochemistry revealed the infiltrate was composed predominantly of CD3+ T lymphocytes (original magnification ×100). B, The enlarged cells were CD4+, consistent with reactive helper T cells (original magnification ×400).

Histopathology—Histologically, AEIs demonstrate a superficial and deep, perivascular, inflammatory infiltrate in the dermis composed of small lymphocytes, some neutrophils, and eosinophils.16 Less common variants of AEI include eosinophilic annular erythema, characterized by a diffuse dermal infiltrate of eosinophils and some lymphocytes, and neutrophilic figurate erythema of infancy, characterized by a dermal infiltrate with neutrophils and leukocytoclasis without vasculitis.1

Our patient’s skin rash was unusual in that the biopsy demonstrated few neutrophils, rare eosinophils, and larger mononuclear cells consistent with reactive helper T lymphocytes. Although these cells may raise concern for an atypical lymphoid infiltrate, recognition of areas with more conventional histopathology of AEIs can facilitate the correct diagnosis.

Differential Diagnosis—The main considerations in the differential diagnosis for AEIs include the following: EAC, familial annular erythema, erythema gyratum atrophicans transiens neonatale, erythema chronicum migrans, urticaria, tinea corporis, neonatal lupus erythematosus, viral exanthems, and leukemia cutis.16

 

 

Erythema annulare centrifugum typically begins in middle age and follows a course of 2 or more years.2 It occurs in association with an underlying infection or neoplasm, and it can develop on the trunk and proximal extremities. Morphologically, EAC can present with arcuate or polycyclic lesions with trailing scale. Histologically, a skin biopsy shows a tight, coat sleeve–like, perivascular, lymphohistiocytic infiltrate in the dermis, with variable epidermal spongiosis and parakeratosis.16 Our patient’s biopsy did show a tight perivascular infiltrate, raising suspicion for EAC. However, the eruption occurred in infancy, and she had no clinical evidence of infection or neoplasm.

Familial annular erythemas can arise within a few days after birth and can present on any part of the body, including the tongue.2 Individual lesions can persist for 4 to 5 days and can accompany congenital malformations. Morphologically, they can present as papules that slowly enlarge to form arcuate lesions with central hyperpigmentation. Histologically, there can be a mild, perivascular, lymphocytic infiltrate in the dermis.16 Our patient’s lesions showed no scale or pigmentation and occurred without a family history or associated malformations.

Erythema gyratum atrophicans transiens neonatale also can arise in the first few days of life and can affect the trunk, neck, and lips.16 Morphologically, the skin lesions can present as arcuate erythematous patches (3–20 mm) with raised borders and central atrophy. Histologically, there is epidermal atrophy with a dermal perivascular mononuclear cell infiltrate with edema. Our patient’s clinical presentation was not classic for this condition, and the lesions showed no atrophy.

Erythema chronicum migrans can arise in children, often with a history of an arthropod bite.13 Morphologically, lesions can evolve over weeks to months and rarely are multiple. Erythema chronicum migrans most commonly occurs in the United States in association with Lyme disease from infection with Borrelia burgdorferi. Histologically, erythema chronicum migrans shows a superficial and deep, perivascular lymphocytic infiltrate in the dermis with plasma cells and eosinophils. A silver stain can demonstrate dermal spirochetes. Our patient had no history of an arthropod bite. A Warthin-Starry stain performed on the biopsy was negative for spirochetes, and serologies for Lyme disease were negative.

Urticaria is rare in neonates and can occur on any part of the body.2 Morphologically, the skin lesions can present as arcuate, erythematous, and polycyclic plaques that wax and wane. Histologically, there is dermal edema with a mild, perivascular and interstitial, mixed inflammatory infiltrate.16 Our patient’s biopsy did not reveal notable edema, and the perivascular infiltrate was coat sleeve–like with few neutrophils and eosinophils. The patient did not respond to initial treatment with antihistamines, making urticaria less likely.

 

 

Tinea corporis is rare in neonates and can occur on any part of the body.13 Morphologically, it can present as scaly annular lesions that are fixed and more persistent. Histologically, there are fungal hyphae and/or yeast in the stratum corneum with spongiotic dermatitis and parakeratosis. Our patient’s lesions were not scaly, and the biopsy demonstrated minimal spongiosis. A periodic acid–Schiff special stain was negative for fungal microorganisms.

Neonatal lupus erythematosus can arise at birth or during the first few weeks of life.16 Morphologically, the skin lesions occur on the scalp, forehead, or neck in a periorbital or malar distribution. They can present as erythematous, annular, scaly patches and plaques. Transplacental transmission of material autoantibodies has been implicated in the etiology, and a complication is infantile heart block. Histologically, a skin biopsy typically shows interface/lichenoid dermatitis. However, our patient’s biopsy did not demonstrate interface changes, and serologically she was negative for autoantibodies.

Viral exanthems are skin eruptions that accompany underlying viral infections.17 Morphologically, patients can present with an erythematous maculopapular rash, sometimes with vesicular, petechial, and urticarial lesions. Laboratory confirmation is made by virus-specific serologies. Histologically, viral exanthems can show a superficial, perivascular, lymphocytic infiltrate in the dermis, with reactive T cells and epidermal spongiosis. Our patient was afebrile and had no known sick contacts. A cytomegalovirus immunohistochemical study on the biopsy was negative, and an Epstein-Barr encoding region in situ hybridization study was negative.

Leukemia cutis is the infiltration of the skin by leukemic cells, most often in conjunction with systemic leukemia.18 In infants and children, the most common leukemia is B-cell acute lymphoblastic leukemia. Morphologically, the skin lesions are characterized by single or multiple violaceous papules, nodules, and plaques. Histologically, there is a perivascular to interstitial infiltrate of atypical mononuclear cells in the dermis and sometimes subcutis. The leukemic cells demonstrate enlarged nuclei with coarse chromatin and prominent nucleoli. Increased mitotic activity may be seen with karyorrhectic debris. Immunohistochemically, the tumor cells can be positive for myeloperoxidase, CD43, CD68, CD34, and CD117.18 Although our patient’s biopsy demonstrated mononuclear cells with karyorrhexis, the cells did not have striking atypia and were negative for blast markers. A recent complete blood cell count on the patient was normal.

Conclusion

We report an unusual case of AEI with mononuclear cells consistent with helper T cells. One must keep these cells in mind when evaluating a biopsy of AEI, as they are benign and not suggestive of an atypical lymphoid infiltrate or leukemia cutis. This will prevent misdiagnosis and ensure that the patient receives appropriate management.

Annular erythemas of infancy (AEIs) are rare benign skin eruptions characterized by annular or circinate, erythematous patches and plaques that arise in patients younger than 1 year.1 Annular erythemas of infancy originally were described by Peterson and Jarratt2 in 1981. Relatively few cases of AEIs have been reported in the literature (eTable).2-15

 

Case Report

An 11-month-old girl presented to dermatology for a rash characterized by annular erythematous patches and plaques on the back, arms, and legs (Figure 1). Three months prior, the rash was more diffuse, monomorphic, and papular. Based on physical examination, the differential diagnosis included a gyrate erythema such as erythema annulare centrifugum (EAC), neonatal lupus, a viral exanthem, leukemia cutis, and AEI. A skin punch biopsy was performed.

FIGURE 1. A, An 11-month-old girl with annular erythematous patches and plaques on the back. B, Annular erythematous lesions were present on the right arm, from which a punch biopsy was taken.

Histologically, the biopsy revealed a superficial to mid dermal, tight, coat sleeve–like, perivascular lymphohistiocytic infiltrate admixed with rare neutrophils in eosinophils within the dermis (Figure 2A). The infiltrate also contained numerous large mononuclear cells with enlarged nuclei, fine loose chromatin, rare nucleoli, and a thin rim of cytoplasm (Figure 2B). There were associated apoptotic bodies with karyorrhectic debris. Immunohistochemistry exhibited enlarged cells that were strong staining with CD3 and CD4, which was consistent with reactive helper T cells (Figure 3). A myeloperoxidase stain highlighted few neutrophils. Stains for terminal deoxynucleotidyl transferase, CD1a, CD117, and CD34 were negative. These findings along with the clinical presentation yielded a diagnosis of AEI with reactive helper T cells.

FIGURE 2. Histopathology demonstrated annular erythema of infancy with mononuclear cells. A, There was a superficial to mid dermal, tight, coat sleeve–like, perivascular, lymphohistiocytic infiltrate admixed with rare neutrophils in eosinophils within the dermis (H&E, original magnification ×40). B, The infiltrate contained numerous large mononuclear cells with enlarged nuclei, fine loose chromatin, rare nucleoli, and a thin rim of cytoplasm (H&E, original magnification ×400).

Comment

Clinical Presentation of AEIs—Annular erythemas of infancy are rare benign skin eruptions that develop in the first few months of life.1,16 Few cases have been reported (eTable). Clinically, AEIs are characterized by annular or circinate, erythematous patches and plaques. They can occur on the face, trunk, and extremities, and they completely resolve by 1 year of age in most cases. One case was reported to persist in a patient from birth until 15 years of age.9 It is thought that AEIs may occur as a hypersensitivity reaction to an unrecognized antigen.

FIGURE 3. A, Immunohistochemistry revealed the infiltrate was composed predominantly of CD3+ T lymphocytes (original magnification ×100). B, The enlarged cells were CD4+, consistent with reactive helper T cells (original magnification ×400).

Histopathology—Histologically, AEIs demonstrate a superficial and deep, perivascular, inflammatory infiltrate in the dermis composed of small lymphocytes, some neutrophils, and eosinophils.16 Less common variants of AEI include eosinophilic annular erythema, characterized by a diffuse dermal infiltrate of eosinophils and some lymphocytes, and neutrophilic figurate erythema of infancy, characterized by a dermal infiltrate with neutrophils and leukocytoclasis without vasculitis.1

Our patient’s skin rash was unusual in that the biopsy demonstrated few neutrophils, rare eosinophils, and larger mononuclear cells consistent with reactive helper T lymphocytes. Although these cells may raise concern for an atypical lymphoid infiltrate, recognition of areas with more conventional histopathology of AEIs can facilitate the correct diagnosis.

Differential Diagnosis—The main considerations in the differential diagnosis for AEIs include the following: EAC, familial annular erythema, erythema gyratum atrophicans transiens neonatale, erythema chronicum migrans, urticaria, tinea corporis, neonatal lupus erythematosus, viral exanthems, and leukemia cutis.16

 

 

Erythema annulare centrifugum typically begins in middle age and follows a course of 2 or more years.2 It occurs in association with an underlying infection or neoplasm, and it can develop on the trunk and proximal extremities. Morphologically, EAC can present with arcuate or polycyclic lesions with trailing scale. Histologically, a skin biopsy shows a tight, coat sleeve–like, perivascular, lymphohistiocytic infiltrate in the dermis, with variable epidermal spongiosis and parakeratosis.16 Our patient’s biopsy did show a tight perivascular infiltrate, raising suspicion for EAC. However, the eruption occurred in infancy, and she had no clinical evidence of infection or neoplasm.

Familial annular erythemas can arise within a few days after birth and can present on any part of the body, including the tongue.2 Individual lesions can persist for 4 to 5 days and can accompany congenital malformations. Morphologically, they can present as papules that slowly enlarge to form arcuate lesions with central hyperpigmentation. Histologically, there can be a mild, perivascular, lymphocytic infiltrate in the dermis.16 Our patient’s lesions showed no scale or pigmentation and occurred without a family history or associated malformations.

Erythema gyratum atrophicans transiens neonatale also can arise in the first few days of life and can affect the trunk, neck, and lips.16 Morphologically, the skin lesions can present as arcuate erythematous patches (3–20 mm) with raised borders and central atrophy. Histologically, there is epidermal atrophy with a dermal perivascular mononuclear cell infiltrate with edema. Our patient’s clinical presentation was not classic for this condition, and the lesions showed no atrophy.

Erythema chronicum migrans can arise in children, often with a history of an arthropod bite.13 Morphologically, lesions can evolve over weeks to months and rarely are multiple. Erythema chronicum migrans most commonly occurs in the United States in association with Lyme disease from infection with Borrelia burgdorferi. Histologically, erythema chronicum migrans shows a superficial and deep, perivascular lymphocytic infiltrate in the dermis with plasma cells and eosinophils. A silver stain can demonstrate dermal spirochetes. Our patient had no history of an arthropod bite. A Warthin-Starry stain performed on the biopsy was negative for spirochetes, and serologies for Lyme disease were negative.

Urticaria is rare in neonates and can occur on any part of the body.2 Morphologically, the skin lesions can present as arcuate, erythematous, and polycyclic plaques that wax and wane. Histologically, there is dermal edema with a mild, perivascular and interstitial, mixed inflammatory infiltrate.16 Our patient’s biopsy did not reveal notable edema, and the perivascular infiltrate was coat sleeve–like with few neutrophils and eosinophils. The patient did not respond to initial treatment with antihistamines, making urticaria less likely.

 

 

Tinea corporis is rare in neonates and can occur on any part of the body.13 Morphologically, it can present as scaly annular lesions that are fixed and more persistent. Histologically, there are fungal hyphae and/or yeast in the stratum corneum with spongiotic dermatitis and parakeratosis. Our patient’s lesions were not scaly, and the biopsy demonstrated minimal spongiosis. A periodic acid–Schiff special stain was negative for fungal microorganisms.

Neonatal lupus erythematosus can arise at birth or during the first few weeks of life.16 Morphologically, the skin lesions occur on the scalp, forehead, or neck in a periorbital or malar distribution. They can present as erythematous, annular, scaly patches and plaques. Transplacental transmission of material autoantibodies has been implicated in the etiology, and a complication is infantile heart block. Histologically, a skin biopsy typically shows interface/lichenoid dermatitis. However, our patient’s biopsy did not demonstrate interface changes, and serologically she was negative for autoantibodies.

Viral exanthems are skin eruptions that accompany underlying viral infections.17 Morphologically, patients can present with an erythematous maculopapular rash, sometimes with vesicular, petechial, and urticarial lesions. Laboratory confirmation is made by virus-specific serologies. Histologically, viral exanthems can show a superficial, perivascular, lymphocytic infiltrate in the dermis, with reactive T cells and epidermal spongiosis. Our patient was afebrile and had no known sick contacts. A cytomegalovirus immunohistochemical study on the biopsy was negative, and an Epstein-Barr encoding region in situ hybridization study was negative.

Leukemia cutis is the infiltration of the skin by leukemic cells, most often in conjunction with systemic leukemia.18 In infants and children, the most common leukemia is B-cell acute lymphoblastic leukemia. Morphologically, the skin lesions are characterized by single or multiple violaceous papules, nodules, and plaques. Histologically, there is a perivascular to interstitial infiltrate of atypical mononuclear cells in the dermis and sometimes subcutis. The leukemic cells demonstrate enlarged nuclei with coarse chromatin and prominent nucleoli. Increased mitotic activity may be seen with karyorrhectic debris. Immunohistochemically, the tumor cells can be positive for myeloperoxidase, CD43, CD68, CD34, and CD117.18 Although our patient’s biopsy demonstrated mononuclear cells with karyorrhexis, the cells did not have striking atypia and were negative for blast markers. A recent complete blood cell count on the patient was normal.

Conclusion

We report an unusual case of AEI with mononuclear cells consistent with helper T cells. One must keep these cells in mind when evaluating a biopsy of AEI, as they are benign and not suggestive of an atypical lymphoid infiltrate or leukemia cutis. This will prevent misdiagnosis and ensure that the patient receives appropriate management.

References
  1. Ríos-Martín JJ, Ferrándiz-Pulido L, Moreno-Ramírez D. Approaches to the dermatopathologic diagnosis of figurate lesions [in Spanish]. Actas Dermosifiliogr. 2011;102:316-324. doi:10.1016/j.ad.2010.12.009
  2. Peterson AO, Jarratt M. Annular erythema of infancy. Arch Dermatol. 1981;117:145-148.
  3. Toonstra J, de Wit RF. “Persistent” annular erythema of infancy. Arch Dermatol.1984;120:1069-1072.
  4. Hebert AA, Esterly NB. Annular erythema of infancy. J Am Acad Dermatol. 1986;14:339-343.
  5. Cox NH, McQueen A, Evans TJ, et al. An annular erythema of infancy. Arch Dermatol. 1987;123:510-513.
  6. Helm TN, Bass J, Chang LW, et al. Persistent annular erythema of infancy. Pediatr Dermatol. 1993;10:46-48.
  7. Kunz M, Hamm K, Bröcker EB, et al. Annular erythema in childhood—a new eosinophilic dermatosis [in German]. Hautarzt. 1998;49:131-134.
  8. Stachowitz S, Abeck D, Schmidt T, et al. Persistent annular erythema of infancy associated with intestinal Candida colonization. Clin Exp Dermatol. 2000;25:404-405.
  9. Wong L-C, Kakakios A, Rogers M. Congenital annular erythema persisting in a 15-year-old girl. Australas J Dermatol. 2002;43:55-61.
  10. Patrizi A, Savoia F, Varotti E, et al. Neutrophilic figurate erythema of infancy. Pediatr Dermatol. 2008;25:255-260. doi:10.1111/j.1525-1470.2008.00646.x
  11. Saha A, Seth J, Mukherjee S, et al. Annular erythema of infancy: a diagnostic challenge. Indian J Paediatr Dermatol. 2014;15:147-149. doi:10.4103/2319-7250.143678
  12. Pfingstler LF, Miller KP, Pride H. Recurring diffuse annular erythematous plaques in a newborn. JAMA Dermatol. 2014;150:565-566. doi:10.1001/jamadermatol.2013.8059
  13. Del Puerto Troncoso C, Curi Tuma M, González Bombardiere S, et al. Neutrophilic figurate erythema of infancy associated with juvenile myelomonocytic leukemia. Actas Dermosifiliogr. 2015;106:431-433. doi:10.1016/j.ad.2014.09.013
  14. Hamidi S, Prose NS, Selim MA. Neutrophilic figurate erythema of infancy: a diagnostic challenge [published online December 26, 2018]. J Cutan Pathol. 2019;46:216-220. doi:10.1111/cup.13394
  15. Patel N, Goldbach H, Hogeling M. An annular eruption in a young child. JAMA Dermatol. 2018;154:1213-1214. doi:10.1001/jamadermatol.2018.1174
  16. Palit A, Inamadar AC. Annular, erythematous skin lesions in a neonate. Indian Dermatol Online J. 2012;3:45-47. doi:10.4103/2229-5178.93504
  17. Keighley CL, Saunderson RB, Kok J, et al. Viral exanthems. Curr Opin Infect Dis. 2015;28:139-150. doi:10.1097/QCO.0000000000000145
  18. Cronin DMP, George TI, Sundram UN. An updated approach to the diagnosis of myeloid leukemia cutis. Am J Clin Pathol. 2009;132:101-110. doi:10.1309/AJCP6GR8BDEXPKHR
References
  1. Ríos-Martín JJ, Ferrándiz-Pulido L, Moreno-Ramírez D. Approaches to the dermatopathologic diagnosis of figurate lesions [in Spanish]. Actas Dermosifiliogr. 2011;102:316-324. doi:10.1016/j.ad.2010.12.009
  2. Peterson AO, Jarratt M. Annular erythema of infancy. Arch Dermatol. 1981;117:145-148.
  3. Toonstra J, de Wit RF. “Persistent” annular erythema of infancy. Arch Dermatol.1984;120:1069-1072.
  4. Hebert AA, Esterly NB. Annular erythema of infancy. J Am Acad Dermatol. 1986;14:339-343.
  5. Cox NH, McQueen A, Evans TJ, et al. An annular erythema of infancy. Arch Dermatol. 1987;123:510-513.
  6. Helm TN, Bass J, Chang LW, et al. Persistent annular erythema of infancy. Pediatr Dermatol. 1993;10:46-48.
  7. Kunz M, Hamm K, Bröcker EB, et al. Annular erythema in childhood—a new eosinophilic dermatosis [in German]. Hautarzt. 1998;49:131-134.
  8. Stachowitz S, Abeck D, Schmidt T, et al. Persistent annular erythema of infancy associated with intestinal Candida colonization. Clin Exp Dermatol. 2000;25:404-405.
  9. Wong L-C, Kakakios A, Rogers M. Congenital annular erythema persisting in a 15-year-old girl. Australas J Dermatol. 2002;43:55-61.
  10. Patrizi A, Savoia F, Varotti E, et al. Neutrophilic figurate erythema of infancy. Pediatr Dermatol. 2008;25:255-260. doi:10.1111/j.1525-1470.2008.00646.x
  11. Saha A, Seth J, Mukherjee S, et al. Annular erythema of infancy: a diagnostic challenge. Indian J Paediatr Dermatol. 2014;15:147-149. doi:10.4103/2319-7250.143678
  12. Pfingstler LF, Miller KP, Pride H. Recurring diffuse annular erythematous plaques in a newborn. JAMA Dermatol. 2014;150:565-566. doi:10.1001/jamadermatol.2013.8059
  13. Del Puerto Troncoso C, Curi Tuma M, González Bombardiere S, et al. Neutrophilic figurate erythema of infancy associated with juvenile myelomonocytic leukemia. Actas Dermosifiliogr. 2015;106:431-433. doi:10.1016/j.ad.2014.09.013
  14. Hamidi S, Prose NS, Selim MA. Neutrophilic figurate erythema of infancy: a diagnostic challenge [published online December 26, 2018]. J Cutan Pathol. 2019;46:216-220. doi:10.1111/cup.13394
  15. Patel N, Goldbach H, Hogeling M. An annular eruption in a young child. JAMA Dermatol. 2018;154:1213-1214. doi:10.1001/jamadermatol.2018.1174
  16. Palit A, Inamadar AC. Annular, erythematous skin lesions in a neonate. Indian Dermatol Online J. 2012;3:45-47. doi:10.4103/2229-5178.93504
  17. Keighley CL, Saunderson RB, Kok J, et al. Viral exanthems. Curr Opin Infect Dis. 2015;28:139-150. doi:10.1097/QCO.0000000000000145
  18. Cronin DMP, George TI, Sundram UN. An updated approach to the diagnosis of myeloid leukemia cutis. Am J Clin Pathol. 2009;132:101-110. doi:10.1309/AJCP6GR8BDEXPKHR
Issue
Cutis - 108(5)
Issue
Cutis - 108(5)
Page Number
289-E2
Page Number
289-E2
Publications
Publications
Topics
Article Type
Display Headline
Annular Erythema of Infancy With Reactive Helper T Lymphocytes
Display Headline
Annular Erythema of Infancy With Reactive Helper T Lymphocytes
Sections
Inside the Article

Practice Points

  • Annular erythemas of infancy (AEIs) are rare benign skin eruptions characterized by persistent, annular, urticarial, nonpruritic patches and plaques that develop in patients younger than 1 year.
  • Although AEIs are benign, lesions with uncommon histologic features such as large mononuclear cells consistent with reactive helper T lymphocytes may pose diagnostic challenges.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Pediatric Subungual Exostosis

Article Type
Changed
Mon, 12/06/2021 - 10:37
Display Headline
Pediatric Subungual Exostosis

Exostosis is a type of benign bone tumor in which trabecular (spongy) bone overgrows its normal border in a nodular pattern. 1,2 Histologically, it usually is surrounded by a fibrocartilaginous cap. 3 It is most commonly found on the lateral or medial aspect of the foot and is thought to be caused by trauma, either physical pressure or infection. 4 When this lesion is found under the nail bed, it is termed subungual exostosis ( Dupuytren exostosis ) . 3 Sequelae of a subungual exostosis include nail dystrophy and lifting of the nail away from the toe, in addition to infection and possible loss of the toenail (onycholysis). There are only 2 genetic conditions related to exostosis: hereditary multiple exostosis and multiple exostoses-mental retardation syndrome.

An exostosis may appear to be a wart on first inspection. It may present similar to osteochondromas, and the only way to get a true diagnosis is by biopsy of the lesion. The treatment for an exostosis is surgery. The surgeon must remove the lesion at the base of the bone from which it grows to prevent recurrence of the lesion.5

Because exostosis may cause nail bed disruption, the differential diagnosis may include nail deformities, such as traumatic onycholysis, onychogryphosis, verrucae, subungual infection, or nail trauma.6,7

 

Case Report

A 7-year-old boy presented with changes of the right great toenail over the last 4 months. The patient noted that the affected nail was discolored, dystrophic, painful, and thickened. He did not recall prior trauma to the affected nail, and his mother stated that the lesion was growing and becoming more painful with a throbbing sensation at times. He described the pain as stabbing, which was exacerbated while walking and playing sports. Neither the patient nor his family had ever had any similar condition. He was not taking any medications, only a daily multivitamin. He had a history of eczematous dermatitis and keratosis pilaris without any other medical illnesses. He had a family history of psoriasis; however, no prior instances of exostosis had been reported. He had no medication allergies.

A full-body cutaneous and nail examination showed a well-developed, well-nourished boy who was in no acute distress. A firm, subungual, pink, pearly,hyperkeratotic nodule was appreciated on the right great toe (Figure 1). The lesion was tender to palpation. The rest of the examination and review of systems were normal.

FIGURE 1. Subungual, pink, pearly nodule on the right great toe.

From the clinical findings, a differential diagnosis of glomus tumor, hemangioma, and infection was considered. Periodic acid–Schiff stain was negative, which ruled out fungal infection. Nail avulsion and a shave biopsy were performed under general anesthesia. There was an exostosis arising from the dorsal aspect of the great toe measuring approximately 5 mm in width at the base and approximately 1 mm in height, which endorsed a diagnosis of distal phalanx subungual exostosis. A postsurgery radiograph (Figure 2) showed residual bone below the level of shave removal at the nail bed.

FIGURE 2.  A radiograph of the right great toe showed soft-tissue changes.

Comment

Exostosis is most commonly found on the lateral or medial aspect of the hallux (great toe) in patients younger than 18 years.8 Diagnosis often is obvious, even without a radiograph or biopsy, because the exostosis comes out from under the tip of the nail. Our case was interesting because the patient was a child, and the exostosis did not lift the nail or extrude from the distal tip of the nail bed. Evidence suggests that a greater-than-expected genetic influence contributes to an exostosis, though further investigation is needed to determine all of the causes and risk factors for subungual bony exostosis. Timely diagnosis and treatment are essential to the prevention of sequelae of the disease, such as toe infection or chronic pain.

References
  1. de Palma L, Gigante A, Specchia N. Subungual exostosis of the foot. Foot Ankle Int. 1996;17:758-763. doi:10.1177/107110079601701208
  2. Multhopp-Stephens H, Walling AK. Subungual (Dupuytren’s) exostosis. J Pediatr Orthop. 1995;15:582-584. doi:10.1097/01241398-199509000-00006
  3. Davis DA, Cohen PR. Subungual exostosis: case report and review of the literature. Pediatr Dermatol. 1996;13:212-218.
  4. Guarneri C, Guarneri F, Risitano G, et al. Solitary asymptomatic nodule of the great toe. Int J Dermatol. 2005;44:245-247.
  5. Letts M, Davidson D, Nizalik E. Subungual exostosis: diagnosis and treatment in children. J Trauma. 1998;44:346-349.
  6. Hoy NY, Leung AKC, Metelitsa AI, et al. New concepts in median nail dystrophy, onychomycosis, and hand, foot, and mouth disease nail pathology. ISRN Dermatol. 2012;2012:680163.
  7. Rich P, Scher RK. Examination of the nail and work-up of nail conditions. In: Rich P, Scher RK, eds. An Atlas of Diseases of the Nail. Parthenon Publishing; 2003.
  8. DaCambra MP, Gupta SK, Ferri-de-Barros F. Subungual exostosis of the toes: a systematic review. Clin Orthop Relat Res. 2014;472:1251-1259. doi:10.1007/s11999-013-3345-4
Article PDF
Author and Disclosure Information

Drs. Yousefian and Davis are from the University of the Incarnate Word School of Osteopathic Medicine, San Antonio, Texas. Dr. Browning is from Texas Dermatology and Laser Specialists, San Antonio.

The authors report no conflict of interest.

Correspondence: Faraz Yousefian, DO, University of the Incarnate Word School of Osteopathic Medicine, 7615-1 Kennedy Circle, San Antonio, TX 78235 ([email protected]).

Issue
Cutis - 108(5)
Publications
Topics
Page Number
256-257
Sections
Author and Disclosure Information

Drs. Yousefian and Davis are from the University of the Incarnate Word School of Osteopathic Medicine, San Antonio, Texas. Dr. Browning is from Texas Dermatology and Laser Specialists, San Antonio.

The authors report no conflict of interest.

Correspondence: Faraz Yousefian, DO, University of the Incarnate Word School of Osteopathic Medicine, 7615-1 Kennedy Circle, San Antonio, TX 78235 ([email protected]).

Author and Disclosure Information

Drs. Yousefian and Davis are from the University of the Incarnate Word School of Osteopathic Medicine, San Antonio, Texas. Dr. Browning is from Texas Dermatology and Laser Specialists, San Antonio.

The authors report no conflict of interest.

Correspondence: Faraz Yousefian, DO, University of the Incarnate Word School of Osteopathic Medicine, 7615-1 Kennedy Circle, San Antonio, TX 78235 ([email protected]).

Article PDF
Article PDF

Exostosis is a type of benign bone tumor in which trabecular (spongy) bone overgrows its normal border in a nodular pattern. 1,2 Histologically, it usually is surrounded by a fibrocartilaginous cap. 3 It is most commonly found on the lateral or medial aspect of the foot and is thought to be caused by trauma, either physical pressure or infection. 4 When this lesion is found under the nail bed, it is termed subungual exostosis ( Dupuytren exostosis ) . 3 Sequelae of a subungual exostosis include nail dystrophy and lifting of the nail away from the toe, in addition to infection and possible loss of the toenail (onycholysis). There are only 2 genetic conditions related to exostosis: hereditary multiple exostosis and multiple exostoses-mental retardation syndrome.

An exostosis may appear to be a wart on first inspection. It may present similar to osteochondromas, and the only way to get a true diagnosis is by biopsy of the lesion. The treatment for an exostosis is surgery. The surgeon must remove the lesion at the base of the bone from which it grows to prevent recurrence of the lesion.5

Because exostosis may cause nail bed disruption, the differential diagnosis may include nail deformities, such as traumatic onycholysis, onychogryphosis, verrucae, subungual infection, or nail trauma.6,7

 

Case Report

A 7-year-old boy presented with changes of the right great toenail over the last 4 months. The patient noted that the affected nail was discolored, dystrophic, painful, and thickened. He did not recall prior trauma to the affected nail, and his mother stated that the lesion was growing and becoming more painful with a throbbing sensation at times. He described the pain as stabbing, which was exacerbated while walking and playing sports. Neither the patient nor his family had ever had any similar condition. He was not taking any medications, only a daily multivitamin. He had a history of eczematous dermatitis and keratosis pilaris without any other medical illnesses. He had a family history of psoriasis; however, no prior instances of exostosis had been reported. He had no medication allergies.

A full-body cutaneous and nail examination showed a well-developed, well-nourished boy who was in no acute distress. A firm, subungual, pink, pearly,hyperkeratotic nodule was appreciated on the right great toe (Figure 1). The lesion was tender to palpation. The rest of the examination and review of systems were normal.

FIGURE 1. Subungual, pink, pearly nodule on the right great toe.

From the clinical findings, a differential diagnosis of glomus tumor, hemangioma, and infection was considered. Periodic acid–Schiff stain was negative, which ruled out fungal infection. Nail avulsion and a shave biopsy were performed under general anesthesia. There was an exostosis arising from the dorsal aspect of the great toe measuring approximately 5 mm in width at the base and approximately 1 mm in height, which endorsed a diagnosis of distal phalanx subungual exostosis. A postsurgery radiograph (Figure 2) showed residual bone below the level of shave removal at the nail bed.

FIGURE 2.  A radiograph of the right great toe showed soft-tissue changes.

Comment

Exostosis is most commonly found on the lateral or medial aspect of the hallux (great toe) in patients younger than 18 years.8 Diagnosis often is obvious, even without a radiograph or biopsy, because the exostosis comes out from under the tip of the nail. Our case was interesting because the patient was a child, and the exostosis did not lift the nail or extrude from the distal tip of the nail bed. Evidence suggests that a greater-than-expected genetic influence contributes to an exostosis, though further investigation is needed to determine all of the causes and risk factors for subungual bony exostosis. Timely diagnosis and treatment are essential to the prevention of sequelae of the disease, such as toe infection or chronic pain.

Exostosis is a type of benign bone tumor in which trabecular (spongy) bone overgrows its normal border in a nodular pattern. 1,2 Histologically, it usually is surrounded by a fibrocartilaginous cap. 3 It is most commonly found on the lateral or medial aspect of the foot and is thought to be caused by trauma, either physical pressure or infection. 4 When this lesion is found under the nail bed, it is termed subungual exostosis ( Dupuytren exostosis ) . 3 Sequelae of a subungual exostosis include nail dystrophy and lifting of the nail away from the toe, in addition to infection and possible loss of the toenail (onycholysis). There are only 2 genetic conditions related to exostosis: hereditary multiple exostosis and multiple exostoses-mental retardation syndrome.

An exostosis may appear to be a wart on first inspection. It may present similar to osteochondromas, and the only way to get a true diagnosis is by biopsy of the lesion. The treatment for an exostosis is surgery. The surgeon must remove the lesion at the base of the bone from which it grows to prevent recurrence of the lesion.5

Because exostosis may cause nail bed disruption, the differential diagnosis may include nail deformities, such as traumatic onycholysis, onychogryphosis, verrucae, subungual infection, or nail trauma.6,7

 

Case Report

A 7-year-old boy presented with changes of the right great toenail over the last 4 months. The patient noted that the affected nail was discolored, dystrophic, painful, and thickened. He did not recall prior trauma to the affected nail, and his mother stated that the lesion was growing and becoming more painful with a throbbing sensation at times. He described the pain as stabbing, which was exacerbated while walking and playing sports. Neither the patient nor his family had ever had any similar condition. He was not taking any medications, only a daily multivitamin. He had a history of eczematous dermatitis and keratosis pilaris without any other medical illnesses. He had a family history of psoriasis; however, no prior instances of exostosis had been reported. He had no medication allergies.

A full-body cutaneous and nail examination showed a well-developed, well-nourished boy who was in no acute distress. A firm, subungual, pink, pearly,hyperkeratotic nodule was appreciated on the right great toe (Figure 1). The lesion was tender to palpation. The rest of the examination and review of systems were normal.

FIGURE 1. Subungual, pink, pearly nodule on the right great toe.

From the clinical findings, a differential diagnosis of glomus tumor, hemangioma, and infection was considered. Periodic acid–Schiff stain was negative, which ruled out fungal infection. Nail avulsion and a shave biopsy were performed under general anesthesia. There was an exostosis arising from the dorsal aspect of the great toe measuring approximately 5 mm in width at the base and approximately 1 mm in height, which endorsed a diagnosis of distal phalanx subungual exostosis. A postsurgery radiograph (Figure 2) showed residual bone below the level of shave removal at the nail bed.

FIGURE 2.  A radiograph of the right great toe showed soft-tissue changes.

Comment

Exostosis is most commonly found on the lateral or medial aspect of the hallux (great toe) in patients younger than 18 years.8 Diagnosis often is obvious, even without a radiograph or biopsy, because the exostosis comes out from under the tip of the nail. Our case was interesting because the patient was a child, and the exostosis did not lift the nail or extrude from the distal tip of the nail bed. Evidence suggests that a greater-than-expected genetic influence contributes to an exostosis, though further investigation is needed to determine all of the causes and risk factors for subungual bony exostosis. Timely diagnosis and treatment are essential to the prevention of sequelae of the disease, such as toe infection or chronic pain.

References
  1. de Palma L, Gigante A, Specchia N. Subungual exostosis of the foot. Foot Ankle Int. 1996;17:758-763. doi:10.1177/107110079601701208
  2. Multhopp-Stephens H, Walling AK. Subungual (Dupuytren’s) exostosis. J Pediatr Orthop. 1995;15:582-584. doi:10.1097/01241398-199509000-00006
  3. Davis DA, Cohen PR. Subungual exostosis: case report and review of the literature. Pediatr Dermatol. 1996;13:212-218.
  4. Guarneri C, Guarneri F, Risitano G, et al. Solitary asymptomatic nodule of the great toe. Int J Dermatol. 2005;44:245-247.
  5. Letts M, Davidson D, Nizalik E. Subungual exostosis: diagnosis and treatment in children. J Trauma. 1998;44:346-349.
  6. Hoy NY, Leung AKC, Metelitsa AI, et al. New concepts in median nail dystrophy, onychomycosis, and hand, foot, and mouth disease nail pathology. ISRN Dermatol. 2012;2012:680163.
  7. Rich P, Scher RK. Examination of the nail and work-up of nail conditions. In: Rich P, Scher RK, eds. An Atlas of Diseases of the Nail. Parthenon Publishing; 2003.
  8. DaCambra MP, Gupta SK, Ferri-de-Barros F. Subungual exostosis of the toes: a systematic review. Clin Orthop Relat Res. 2014;472:1251-1259. doi:10.1007/s11999-013-3345-4
References
  1. de Palma L, Gigante A, Specchia N. Subungual exostosis of the foot. Foot Ankle Int. 1996;17:758-763. doi:10.1177/107110079601701208
  2. Multhopp-Stephens H, Walling AK. Subungual (Dupuytren’s) exostosis. J Pediatr Orthop. 1995;15:582-584. doi:10.1097/01241398-199509000-00006
  3. Davis DA, Cohen PR. Subungual exostosis: case report and review of the literature. Pediatr Dermatol. 1996;13:212-218.
  4. Guarneri C, Guarneri F, Risitano G, et al. Solitary asymptomatic nodule of the great toe. Int J Dermatol. 2005;44:245-247.
  5. Letts M, Davidson D, Nizalik E. Subungual exostosis: diagnosis and treatment in children. J Trauma. 1998;44:346-349.
  6. Hoy NY, Leung AKC, Metelitsa AI, et al. New concepts in median nail dystrophy, onychomycosis, and hand, foot, and mouth disease nail pathology. ISRN Dermatol. 2012;2012:680163.
  7. Rich P, Scher RK. Examination of the nail and work-up of nail conditions. In: Rich P, Scher RK, eds. An Atlas of Diseases of the Nail. Parthenon Publishing; 2003.
  8. DaCambra MP, Gupta SK, Ferri-de-Barros F. Subungual exostosis of the toes: a systematic review. Clin Orthop Relat Res. 2014;472:1251-1259. doi:10.1007/s11999-013-3345-4
Issue
Cutis - 108(5)
Issue
Cutis - 108(5)
Page Number
256-257
Page Number
256-257
Publications
Publications
Topics
Article Type
Display Headline
Pediatric Subungual Exostosis
Display Headline
Pediatric Subungual Exostosis
Sections
Inside the Article

Practice Points

  • Nail dystrophy can have a variety of causes, most commonly trauma, onychomycosis, verrucae, or subungual exostosis.
  • Exostosis is a benign osteochondral tumor commonly found on the lateral or medial aspect of the hallux (great toe) in pediatric and young adult patients.
  • A radiograph can be used as a preliminary tool for diagnosis, but subungual exostosis must be confirmed by biopsy or tissue histology at the time of excision.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Early Pilomatrix Carcinoma: A Case Report With Emphasis on Molecular Pathology and Review of the Literature

Article Type
Changed
Thu, 11/04/2021 - 09:20
Display Headline
Early Pilomatrix Carcinoma: A Case Report With Emphasis on Molecular Pathology and Review of the Literature

Pilomatrix carcinoma is a rare adnexal tumor with origin from the germinative matrical cells of the hair follicle. Clinically, it presents as a solitary lesion commonly found in the head and neck region as well as the upper back. The tumors cannot be distinguished by their clinical appearance only and frequently are mistaken for cysts. Histopathologic examination provides the definitive diagnosis in most cases. These carcinomas are aggressive neoplasms with a high probability of local recurrence and distant metastasis. Assessment of the Wnt signaling pathway components such as β-catenin, lymphoid enhancer-binding factor 1 (LEF-1), and caudal-related homeobox transcription factor 2 (CDX-2) potentially can be used for diagnostic purposes and targeted therapy.

We report a rare and unique case of early pilomatrix carcinoma with intralesional melanocytes. We review the molecular pathology and pathogenesis of these carcinomas as well as the significance of early diagnosis.

Case Report

A 73-year-old man with a history of extensive sun exposure presented with a 1-cm, raised, rapidly growing, slightly irregular, purple lesion on the right forearm of 3 months’ duration with tendency to bleed. He did not have a history of skin cancers and was otherwise healthy. Excision was recommended due to the progressive and rapid growth of the lesion.

Histopathologic Findings—Gross examination revealed a 0.9×0.7-cm, raised, slightly irregular lesion located 1 mm away from the closest peripheral margin. Histologically, the lesion was a relatively circumscribed, dermal-based basaloid neoplasm with slightly ill-defined edges involving the superficial and deep dermis (Figure 1A). The neoplasm was formed predominantly of sheets of basaloid cells and small nests of ghost cells, in addition to some squamoid and transitional cells (Figure 1B). The basaloid cells exhibited severe nuclear atypia, pleomorphism, increased nuclear to cytoplasmic ratio (Figure 1C), minimal to moderate amounts of eosinophilic cytoplasm, enlarged nuclei, prominent nucleoli, and coarse chromatin pattern. Abundant mitotic activity and apoptotic bodies were present as well as focal area of central necrosis (Figure 1C). Also, melanophages and a multinucleated giant cell reaction was noted. Elastic trichrome special stain highlighted focal infiltration of the neoplastic cells into the adjacent desmoplastic stroma. Melanin stain was negative for melanin pigment within the neoplasm. Given the presence of severely atypical basaloid cells along with ghost cells indicating matrical differentiation, a diagnosis of pilomatrix carcinoma was rendered.

FIGURE 1. A, Histopathology of a pilomatrix carcinoma revealed a dermal-based neoplasm with irregular borders formed predominantly of basaloid cells (H&E, original magnification ×20) (reference bar, 2 mm). B, The neoplasm was formed of basaloid shadow cells (red stars) and squamoid cells (H&E, original magnification ×200) (reference bar, 200 μm). C, Marked cytologic atypia of basaloid cells with increased mitoses and focal necrosis also were present (H&E, original magnification ×200) (reference bar, 200 μm).

Immunohistochemistry—The neoplastic cells were diffusely positive for p63, CDX-2 (Figure 2A), β-catenin (Figure 2B), and CD10 (Figure 2C), and focally and weakly positive for cytokeratin (CK) 5, BerEP4 (staining the tumor periphery), androgen receptor, and CK18 (a low-molecular-weight keratin). They were negative for monoclonal carcinoembryonic antigen, epithelial membrane antigen, CK7, CK20, CD34, SOX-10, CD56, synaptophysin, and chromogranin. Cytokeratin 14 was positive in the squamoid cells but negative in the basaloid cells. SOX-10 and melanoma cocktail immunostains demonstrated few intralesional dendritic melanocytes.

FIGURE 2. A–C, Immunohistochemistry revealed the tumor cells were positive for caudal-related homeobox transcription factor 2, β-catenin, and CD10 (original magnifications ×40, ×20, and ×20, respectively) (reference bars: 600 μm, 2 mm, and 2 mm, respectively).

Comment

Pilomatrix carcinoma is a rare malignant cutaneous adnexal neoplasm with origin from the germinative matrix of the hair bulb region of hair follicles. Pilomatrix carcinoma was first reported in 1980.1,2 These tumors are characterized by rapid growth and aggressive behavior. Their benign counterpart, pilomatrixoma, is a slow-growing, dermal or subcutaneous tumor that rarely recurs after complete excision.

As with pilomatrixoma, pilomatrix carcinomas are asymptomatic and present as solitary dermal or subcutaneous masses3,4 that most commonly are found in the posterior neck, upper back, and preauricular regions of middle-aged or elderly adults with male predominance.5 They range in size from 0.5 to 20 cm with a mean of 4 cm that is slightly larger than pilomatrixoma. Pilomatrix carcinomas predominantly are firm tumors with or without cystic components, and they exhibit a high probability of recurrence and have risk for distant metastasis.6-15

 

 

The differential diagnosis includes epidermal cysts, pilomatrixoma, basal cell carcinoma with matrical differentiation, trichoblastoma/trichoblastic carcinoma, and trichilemmal carcinoma. Pilomatrix carcinomas frequently are mistaken for epidermal cysts on clinical examination. Such a distinction can be easily resolved by histopathologic evaluation. The more challenging differential diagnosis is with pilomatrixoma. Histologically, pilomatrixomas consist of a distinct population of cells including basaloid, squamoid, transitional, and shadow cells in variable proportions. The basaloid cells transition to shadow cells in an organized zonal fashion.16 Compared to pilomatrixomas, pilomatrix carcinomas often show predominance of the basaloid cells; marked cytologic atypia and pleomorphism; numerous mitotic figures; deep infiltrative pattern into subcutaneous fat, fascia, and skeletal muscle; stromal desmoplasia; necrosis; and neurovascular invasion (Tables 1 and 2). Furthermore, the shadow cells tend to form a small nested pattern in pilomatrix carcinoma instead of the flat sheetlike pattern usually observed in pilomatrixoma.16 Basal cell carcinoma with matrical differentiation can pose a diagnostic challenge in the differential diagnosis; basal cell carcinoma usually exhibits a peripheral palisade of the basaloid cells accompanied by retraction spaces separating the tumor from the stroma. Trichoblastoma/trichoblastic carcinoma with matrical differentiation can be distinguished by its exuberant stroma, prominent primitive hair follicles, and papillary mesenchymal bodies. Trichilemmal carcinomas are recognized by their connection to the overlying epidermis, peripheral palisading, and presence of clear cells, while pilomatrix carcinoma lacks connection to the surface epithelium.

Immunohistochemical stains have little to no role in the differential diagnosis, and morphology is the mainstay in making the diagnosis. Rarely, pilomatrix carcinoma can be confused with poorly differentiated sebaceous carcinoma and poorly differentiated squamous cell carcinoma. Although careful scrutiny of the histologic features may help identify mature sebocytes in sebaceous carcinoma, evidence of keratinization in squamous cell carcinoma and ghost cells in pilomatrix carcinoma, using a panel of immunohistochemical stains can be helpful in reaching the final diagnosis (Table 3).

The development of hair matrix tumors have been known to harbor mutations in exon 3 of the catenin beta-1 gene, CTNNB1, that encodes for β-catenin, a downstream effector in the Wnt signaling pathway responsible for differentiation, proliferation, and adhesion of epithelial stem cells.17-21 In a study conducted by Kazakov et al,22 DNA was extracted from 86 lesions: 4 were pilomatrixomas and 1 was a pilomatrix carcinoma. A polymerase chain reaction assay revealed 8 pathogenic variants of the β-catenin gene. D32Y (CTNNB1):c.94G>T (p.Asp32Tyr) and G34R (CTNNB1):c.100G>C (p.Gly34Arg) were the mutations present in pilomatrixoma and pilomatrix carcinoma, respectively.22 In addition, there are several proteins that are part of the Wnt pathway in addition to β-catenin—LEF-1 and CDX-2.

Tumminello and Hosler23 found that pilomatrixomas and pilomatrix carcinomas were positive for CDX-2, β-catenin, and LEF-1 by immunohistochemistry. These downstream molecules in the Wnt signaling pathway could have the potential to be used as diagnostic and prognostic markers.2,13,15,23

Although the pathogenesis is unclear, there are 2 possible mechanisms by which pilomatrix carcinomas develop. They can either arise as de novo tumors, or it is possible that initial mutations in β-catenin result in the formation of pilomatrixomas at an early age that may undergo malignant transformation in elderly patients over time with additional mutations.2

 

 

Our case was strongly and diffusely positive for β-catenin in a nuclear and cytoplasmic pattern and CDX-2 in a nuclear pattern, supporting the role of the Wnt signaling pathway in such tumors. Furthermore, our case demonstrated the presence of few intralesional normal dendritic melanocytes, a rare finding1,24,25 but not unexpected, as melanocytes normally are present within the hair follicle matrix.

Pilomatrix carcinomas are aggressive tumors with a high risk for local recurrence and tendency for metastasis. In a study of 13 cases of pilomatrix carcinomas, Herrmann et al13 found that metastasis was significantly associated with local tumor recurrence (P<.0413). They concluded that the combination of overall high local recurrence and metastatic rates of pilomatrix carcinoma as well as documented tumor-related deaths would warrant continued patient follow-up, especially for recurrent tumors.13 Rapid growth of a tumor, either de novo or following several months of stable size, should alert physicians to perform a diagnostic biopsy.

Management options of pilomatrix carcinoma include surgery or radiation with close follow-up. The most widely reported treatment of pilomatrix carcinoma is wide local excision with histologically confirmed clear margins. Mohs micrographic surgery is an excellent treatment option.2,13-15 Adjuvant radiation therapy may be necessary following excision. Currently there is no consensus on surgical management, and standard excisional margins have not been defined.26 Jones et al2 concluded that complete excision with wide margins likely is curative, with decreased rates of recurrence, and better awareness of this carcinoma would lead to appropriate treatment while avoiding unnecessary diagnostic tests.2

 

Conclusion

We report an exceptionally unique case of early pilomatrix carcinoma with a discussion on the pathogenesis and molecular pathology of hair matrix tumors. A large cohort of patients with longer follow-up periods and better molecular characterization is essential in drawing accurate information about their prognosis, identifying molecular markers that can be used as therapeutic targets, and determining ideal management strategy.

References
  1. Jani P, Chetty R, Ghazarian DM. An unusual composite pilomatrix carcinoma with intralesional melanocytes: differential diagnosis, immunohistochemical evaluation, and review of the literature. Am J Dermatopathol. 2008;30:174-177.
  2. Jones C, Twoon M, Ho W, et al. Pilomatrix carcinoma: 12-year experience and review of the literature. J Cutan Pathol. 2018;45:33-38.
  3. Forbis R, Helwig EB. Pilomatrixoma (calcifying epithelioma). Arch Dermatol. 1961;83:606.
  4. Elder D, Elenitsas R, Ragsdale BD. Tumors of epidermal appendages. In: Elder D, Elenitsas R, Jaworsky C, eds. Lever’s Histopathology of the Skin. 8th ed. Lippincott Raven; 1997:757-759.
  5. Aherne NJ, Fitzpatrick DA, Gibbons D, et al. Pilomatrix carcinoma presenting as an extra axial mass: clinicopathological features. Diagn Pathol. 2008;3:47.
  6. Papadakis M, de Bree E, Floros N, et al. Pilomatrix carcinoma: more malignant biological behavior than was considered in the past. Mol Clin Oncol. 2017;6:415-418.
  7. LeBoit PE, Parslow TG, Choy SH. Hair matrix differentiation: occurrence in lesions other than pilomatricoma. Am J Dermatopathol. 1987;9:399-405.
  8. Campoy F, Stiefel P, Stiefel E, et al. Pilomatrix carcinoma: role played by MR imaging. Neuroradiology. 1989;31:196-198.
  9. Tateyama H, Eimoto T, Tada T, et al. Malignant pilomatricoma: an immunohistochemical study with antihair keratin antibody. Cancer. 1992;69:127-132.
  10. O’Donovan DG, Freemont AJ, Adams JE, et al. Malignant pilomatrixoma with bone metastasis. Histopathology. 1993;23:385-386.
  11. Cross P, Richmond I, Wells S, et al. Malignant pilomatrixoma with bone metastasis. Histopathology. 1994;24:499-500.
  12. Niedermeyer HP, Peris K, Höfler H. Pilomatrix carcinoma with multiple visceral metastases: report of a case. Cancer. 1996;77:1311-1314.
  13. Herrmann JL, Allan A, Trapp KM, et al. Pilomatrix carcinoma: 13 new cases and review of the literature with emphasis on predictors of metastasis. J Am Acad Dermatol. 2014;71:38-43.
  14. Xing L, Marzolf SA, Vandergriff T, et al. Facial pilomatrix carcinomas treated with Mohs micrographic surgery. JAAD Case Rep. 2018;4:253-255.
  15. Fernandez-Flores A, Cassarino DS. Sarcomatoid pilomatrix carcinoma. J Cutan Pathol. 2018;45:508-514.
  16. Sau P, Lupton GP, Graham JH. Pilomatrix carcinoma. Cancer. 1993;71:2491-2498.
  17. Chan E, Gat U, McNiff JM, et al. A common human skin tumour is caused by activating mutations in β-catenin. Nat Genet. 1999;21:410-413.
  18. Huelsken J, Vogel R, Erdmann B, et al. β-catenin controls hair follicle morphogenesis and stem cell differentiation in the skin. Cell. 2001;105:533-545.
  19. Kikuchi A. Tumor formation by genetic mutations in the components of the Wnt signaling pathway. Cancer Sci. 2003;94:225-229.
  20. Durand M, Moles J. Beta-catenin mutations in a common skin cancer: pilomatricoma. Bull Cancer. 1999;86:725-726.
  21. Lazar AJF, Calonje E, Grayson W, et al. Pilomatrix carcinomas contain mutations in CTNNB1, the gene encoding beta-catenin. J Cutan Pathol. 2005;32:148-157.
  22. Kazakov DV, Sima R, Vanecek T, et al. Mutations in exon 3 of the CTNNB1 gene (β-catenin gene) in cutaneous adnexal tumors. Am J Dermatopathol. 2009;31:248-255.
  23. Tumminello K, Hosler GA. CDX2 and LEF-1 expression in pilomatrical tumors and their utility in the diagnosis of pilomatrical carcinoma. J Cutan Pathol. 2018;45:318-324.
  24. Rodic´ N, Taube JM, Manson P, et al Locally invasive dermal squamomelanocytic tumor with matrical differentiation: a peculiar case with review of the literature. Am J Dermatopathol. 2013;35:E72-E76.
  25. Perez C, Debbaneh M, Cassarino D. Preference for the term pilomatrical carcinoma with melanocytic hyperplasia: letter to the editor. J Cutan Pathol. 2017;44:655-657.
  26. Herrmann JL, Allan A, Trapp KM, et al. Pilomatrix carcinoma: 13 new cases and review of the literature with emphasis on predictors of metastasis. J Am Acad Dermatol. 2014;71:38-43.
Article PDF
Author and Disclosure Information

Drs. Kamil and Ghazarian and Mr. Sachdeva are from the University of Toronto, Ontario, Canada. Drs. Kamil and Ghazarian are from the Department of Laboratory Medicine and Pathobiology. Drs. Kamil and Ghazarian also are from the Department of Laboratory Medicine and Pathobiology, University Health Network, Toronto. Dr. Kwapis is from Sensenbrenner Hospital, Kapuskasing, Ontario, and McMaster University, Hamilton, Ontario.

The authors report no conflict of interest.

Correspondence: Zaid Saeed Kamil, MD, 200 Elizabeth St, Toronto, ON M5G 2C4, Canada ([email protected]).

Issue
Cutis - 108(4)
Publications
Topics
Page Number
E24-E28
Sections
Author and Disclosure Information

Drs. Kamil and Ghazarian and Mr. Sachdeva are from the University of Toronto, Ontario, Canada. Drs. Kamil and Ghazarian are from the Department of Laboratory Medicine and Pathobiology. Drs. Kamil and Ghazarian also are from the Department of Laboratory Medicine and Pathobiology, University Health Network, Toronto. Dr. Kwapis is from Sensenbrenner Hospital, Kapuskasing, Ontario, and McMaster University, Hamilton, Ontario.

The authors report no conflict of interest.

Correspondence: Zaid Saeed Kamil, MD, 200 Elizabeth St, Toronto, ON M5G 2C4, Canada ([email protected]).

Author and Disclosure Information

Drs. Kamil and Ghazarian and Mr. Sachdeva are from the University of Toronto, Ontario, Canada. Drs. Kamil and Ghazarian are from the Department of Laboratory Medicine and Pathobiology. Drs. Kamil and Ghazarian also are from the Department of Laboratory Medicine and Pathobiology, University Health Network, Toronto. Dr. Kwapis is from Sensenbrenner Hospital, Kapuskasing, Ontario, and McMaster University, Hamilton, Ontario.

The authors report no conflict of interest.

Correspondence: Zaid Saeed Kamil, MD, 200 Elizabeth St, Toronto, ON M5G 2C4, Canada ([email protected]).

Article PDF
Article PDF

Pilomatrix carcinoma is a rare adnexal tumor with origin from the germinative matrical cells of the hair follicle. Clinically, it presents as a solitary lesion commonly found in the head and neck region as well as the upper back. The tumors cannot be distinguished by their clinical appearance only and frequently are mistaken for cysts. Histopathologic examination provides the definitive diagnosis in most cases. These carcinomas are aggressive neoplasms with a high probability of local recurrence and distant metastasis. Assessment of the Wnt signaling pathway components such as β-catenin, lymphoid enhancer-binding factor 1 (LEF-1), and caudal-related homeobox transcription factor 2 (CDX-2) potentially can be used for diagnostic purposes and targeted therapy.

We report a rare and unique case of early pilomatrix carcinoma with intralesional melanocytes. We review the molecular pathology and pathogenesis of these carcinomas as well as the significance of early diagnosis.

Case Report

A 73-year-old man with a history of extensive sun exposure presented with a 1-cm, raised, rapidly growing, slightly irregular, purple lesion on the right forearm of 3 months’ duration with tendency to bleed. He did not have a history of skin cancers and was otherwise healthy. Excision was recommended due to the progressive and rapid growth of the lesion.

Histopathologic Findings—Gross examination revealed a 0.9×0.7-cm, raised, slightly irregular lesion located 1 mm away from the closest peripheral margin. Histologically, the lesion was a relatively circumscribed, dermal-based basaloid neoplasm with slightly ill-defined edges involving the superficial and deep dermis (Figure 1A). The neoplasm was formed predominantly of sheets of basaloid cells and small nests of ghost cells, in addition to some squamoid and transitional cells (Figure 1B). The basaloid cells exhibited severe nuclear atypia, pleomorphism, increased nuclear to cytoplasmic ratio (Figure 1C), minimal to moderate amounts of eosinophilic cytoplasm, enlarged nuclei, prominent nucleoli, and coarse chromatin pattern. Abundant mitotic activity and apoptotic bodies were present as well as focal area of central necrosis (Figure 1C). Also, melanophages and a multinucleated giant cell reaction was noted. Elastic trichrome special stain highlighted focal infiltration of the neoplastic cells into the adjacent desmoplastic stroma. Melanin stain was negative for melanin pigment within the neoplasm. Given the presence of severely atypical basaloid cells along with ghost cells indicating matrical differentiation, a diagnosis of pilomatrix carcinoma was rendered.

FIGURE 1. A, Histopathology of a pilomatrix carcinoma revealed a dermal-based neoplasm with irregular borders formed predominantly of basaloid cells (H&E, original magnification ×20) (reference bar, 2 mm). B, The neoplasm was formed of basaloid shadow cells (red stars) and squamoid cells (H&E, original magnification ×200) (reference bar, 200 μm). C, Marked cytologic atypia of basaloid cells with increased mitoses and focal necrosis also were present (H&E, original magnification ×200) (reference bar, 200 μm).

Immunohistochemistry—The neoplastic cells were diffusely positive for p63, CDX-2 (Figure 2A), β-catenin (Figure 2B), and CD10 (Figure 2C), and focally and weakly positive for cytokeratin (CK) 5, BerEP4 (staining the tumor periphery), androgen receptor, and CK18 (a low-molecular-weight keratin). They were negative for monoclonal carcinoembryonic antigen, epithelial membrane antigen, CK7, CK20, CD34, SOX-10, CD56, synaptophysin, and chromogranin. Cytokeratin 14 was positive in the squamoid cells but negative in the basaloid cells. SOX-10 and melanoma cocktail immunostains demonstrated few intralesional dendritic melanocytes.

FIGURE 2. A–C, Immunohistochemistry revealed the tumor cells were positive for caudal-related homeobox transcription factor 2, β-catenin, and CD10 (original magnifications ×40, ×20, and ×20, respectively) (reference bars: 600 μm, 2 mm, and 2 mm, respectively).

Comment

Pilomatrix carcinoma is a rare malignant cutaneous adnexal neoplasm with origin from the germinative matrix of the hair bulb region of hair follicles. Pilomatrix carcinoma was first reported in 1980.1,2 These tumors are characterized by rapid growth and aggressive behavior. Their benign counterpart, pilomatrixoma, is a slow-growing, dermal or subcutaneous tumor that rarely recurs after complete excision.

As with pilomatrixoma, pilomatrix carcinomas are asymptomatic and present as solitary dermal or subcutaneous masses3,4 that most commonly are found in the posterior neck, upper back, and preauricular regions of middle-aged or elderly adults with male predominance.5 They range in size from 0.5 to 20 cm with a mean of 4 cm that is slightly larger than pilomatrixoma. Pilomatrix carcinomas predominantly are firm tumors with or without cystic components, and they exhibit a high probability of recurrence and have risk for distant metastasis.6-15

 

 

The differential diagnosis includes epidermal cysts, pilomatrixoma, basal cell carcinoma with matrical differentiation, trichoblastoma/trichoblastic carcinoma, and trichilemmal carcinoma. Pilomatrix carcinomas frequently are mistaken for epidermal cysts on clinical examination. Such a distinction can be easily resolved by histopathologic evaluation. The more challenging differential diagnosis is with pilomatrixoma. Histologically, pilomatrixomas consist of a distinct population of cells including basaloid, squamoid, transitional, and shadow cells in variable proportions. The basaloid cells transition to shadow cells in an organized zonal fashion.16 Compared to pilomatrixomas, pilomatrix carcinomas often show predominance of the basaloid cells; marked cytologic atypia and pleomorphism; numerous mitotic figures; deep infiltrative pattern into subcutaneous fat, fascia, and skeletal muscle; stromal desmoplasia; necrosis; and neurovascular invasion (Tables 1 and 2). Furthermore, the shadow cells tend to form a small nested pattern in pilomatrix carcinoma instead of the flat sheetlike pattern usually observed in pilomatrixoma.16 Basal cell carcinoma with matrical differentiation can pose a diagnostic challenge in the differential diagnosis; basal cell carcinoma usually exhibits a peripheral palisade of the basaloid cells accompanied by retraction spaces separating the tumor from the stroma. Trichoblastoma/trichoblastic carcinoma with matrical differentiation can be distinguished by its exuberant stroma, prominent primitive hair follicles, and papillary mesenchymal bodies. Trichilemmal carcinomas are recognized by their connection to the overlying epidermis, peripheral palisading, and presence of clear cells, while pilomatrix carcinoma lacks connection to the surface epithelium.

Immunohistochemical stains have little to no role in the differential diagnosis, and morphology is the mainstay in making the diagnosis. Rarely, pilomatrix carcinoma can be confused with poorly differentiated sebaceous carcinoma and poorly differentiated squamous cell carcinoma. Although careful scrutiny of the histologic features may help identify mature sebocytes in sebaceous carcinoma, evidence of keratinization in squamous cell carcinoma and ghost cells in pilomatrix carcinoma, using a panel of immunohistochemical stains can be helpful in reaching the final diagnosis (Table 3).

The development of hair matrix tumors have been known to harbor mutations in exon 3 of the catenin beta-1 gene, CTNNB1, that encodes for β-catenin, a downstream effector in the Wnt signaling pathway responsible for differentiation, proliferation, and adhesion of epithelial stem cells.17-21 In a study conducted by Kazakov et al,22 DNA was extracted from 86 lesions: 4 were pilomatrixomas and 1 was a pilomatrix carcinoma. A polymerase chain reaction assay revealed 8 pathogenic variants of the β-catenin gene. D32Y (CTNNB1):c.94G>T (p.Asp32Tyr) and G34R (CTNNB1):c.100G>C (p.Gly34Arg) were the mutations present in pilomatrixoma and pilomatrix carcinoma, respectively.22 In addition, there are several proteins that are part of the Wnt pathway in addition to β-catenin—LEF-1 and CDX-2.

Tumminello and Hosler23 found that pilomatrixomas and pilomatrix carcinomas were positive for CDX-2, β-catenin, and LEF-1 by immunohistochemistry. These downstream molecules in the Wnt signaling pathway could have the potential to be used as diagnostic and prognostic markers.2,13,15,23

Although the pathogenesis is unclear, there are 2 possible mechanisms by which pilomatrix carcinomas develop. They can either arise as de novo tumors, or it is possible that initial mutations in β-catenin result in the formation of pilomatrixomas at an early age that may undergo malignant transformation in elderly patients over time with additional mutations.2

 

 

Our case was strongly and diffusely positive for β-catenin in a nuclear and cytoplasmic pattern and CDX-2 in a nuclear pattern, supporting the role of the Wnt signaling pathway in such tumors. Furthermore, our case demonstrated the presence of few intralesional normal dendritic melanocytes, a rare finding1,24,25 but not unexpected, as melanocytes normally are present within the hair follicle matrix.

Pilomatrix carcinomas are aggressive tumors with a high risk for local recurrence and tendency for metastasis. In a study of 13 cases of pilomatrix carcinomas, Herrmann et al13 found that metastasis was significantly associated with local tumor recurrence (P<.0413). They concluded that the combination of overall high local recurrence and metastatic rates of pilomatrix carcinoma as well as documented tumor-related deaths would warrant continued patient follow-up, especially for recurrent tumors.13 Rapid growth of a tumor, either de novo or following several months of stable size, should alert physicians to perform a diagnostic biopsy.

Management options of pilomatrix carcinoma include surgery or radiation with close follow-up. The most widely reported treatment of pilomatrix carcinoma is wide local excision with histologically confirmed clear margins. Mohs micrographic surgery is an excellent treatment option.2,13-15 Adjuvant radiation therapy may be necessary following excision. Currently there is no consensus on surgical management, and standard excisional margins have not been defined.26 Jones et al2 concluded that complete excision with wide margins likely is curative, with decreased rates of recurrence, and better awareness of this carcinoma would lead to appropriate treatment while avoiding unnecessary diagnostic tests.2

 

Conclusion

We report an exceptionally unique case of early pilomatrix carcinoma with a discussion on the pathogenesis and molecular pathology of hair matrix tumors. A large cohort of patients with longer follow-up periods and better molecular characterization is essential in drawing accurate information about their prognosis, identifying molecular markers that can be used as therapeutic targets, and determining ideal management strategy.

Pilomatrix carcinoma is a rare adnexal tumor with origin from the germinative matrical cells of the hair follicle. Clinically, it presents as a solitary lesion commonly found in the head and neck region as well as the upper back. The tumors cannot be distinguished by their clinical appearance only and frequently are mistaken for cysts. Histopathologic examination provides the definitive diagnosis in most cases. These carcinomas are aggressive neoplasms with a high probability of local recurrence and distant metastasis. Assessment of the Wnt signaling pathway components such as β-catenin, lymphoid enhancer-binding factor 1 (LEF-1), and caudal-related homeobox transcription factor 2 (CDX-2) potentially can be used for diagnostic purposes and targeted therapy.

We report a rare and unique case of early pilomatrix carcinoma with intralesional melanocytes. We review the molecular pathology and pathogenesis of these carcinomas as well as the significance of early diagnosis.

Case Report

A 73-year-old man with a history of extensive sun exposure presented with a 1-cm, raised, rapidly growing, slightly irregular, purple lesion on the right forearm of 3 months’ duration with tendency to bleed. He did not have a history of skin cancers and was otherwise healthy. Excision was recommended due to the progressive and rapid growth of the lesion.

Histopathologic Findings—Gross examination revealed a 0.9×0.7-cm, raised, slightly irregular lesion located 1 mm away from the closest peripheral margin. Histologically, the lesion was a relatively circumscribed, dermal-based basaloid neoplasm with slightly ill-defined edges involving the superficial and deep dermis (Figure 1A). The neoplasm was formed predominantly of sheets of basaloid cells and small nests of ghost cells, in addition to some squamoid and transitional cells (Figure 1B). The basaloid cells exhibited severe nuclear atypia, pleomorphism, increased nuclear to cytoplasmic ratio (Figure 1C), minimal to moderate amounts of eosinophilic cytoplasm, enlarged nuclei, prominent nucleoli, and coarse chromatin pattern. Abundant mitotic activity and apoptotic bodies were present as well as focal area of central necrosis (Figure 1C). Also, melanophages and a multinucleated giant cell reaction was noted. Elastic trichrome special stain highlighted focal infiltration of the neoplastic cells into the adjacent desmoplastic stroma. Melanin stain was negative for melanin pigment within the neoplasm. Given the presence of severely atypical basaloid cells along with ghost cells indicating matrical differentiation, a diagnosis of pilomatrix carcinoma was rendered.

FIGURE 1. A, Histopathology of a pilomatrix carcinoma revealed a dermal-based neoplasm with irregular borders formed predominantly of basaloid cells (H&E, original magnification ×20) (reference bar, 2 mm). B, The neoplasm was formed of basaloid shadow cells (red stars) and squamoid cells (H&E, original magnification ×200) (reference bar, 200 μm). C, Marked cytologic atypia of basaloid cells with increased mitoses and focal necrosis also were present (H&E, original magnification ×200) (reference bar, 200 μm).

Immunohistochemistry—The neoplastic cells were diffusely positive for p63, CDX-2 (Figure 2A), β-catenin (Figure 2B), and CD10 (Figure 2C), and focally and weakly positive for cytokeratin (CK) 5, BerEP4 (staining the tumor periphery), androgen receptor, and CK18 (a low-molecular-weight keratin). They were negative for monoclonal carcinoembryonic antigen, epithelial membrane antigen, CK7, CK20, CD34, SOX-10, CD56, synaptophysin, and chromogranin. Cytokeratin 14 was positive in the squamoid cells but negative in the basaloid cells. SOX-10 and melanoma cocktail immunostains demonstrated few intralesional dendritic melanocytes.

FIGURE 2. A–C, Immunohistochemistry revealed the tumor cells were positive for caudal-related homeobox transcription factor 2, β-catenin, and CD10 (original magnifications ×40, ×20, and ×20, respectively) (reference bars: 600 μm, 2 mm, and 2 mm, respectively).

Comment

Pilomatrix carcinoma is a rare malignant cutaneous adnexal neoplasm with origin from the germinative matrix of the hair bulb region of hair follicles. Pilomatrix carcinoma was first reported in 1980.1,2 These tumors are characterized by rapid growth and aggressive behavior. Their benign counterpart, pilomatrixoma, is a slow-growing, dermal or subcutaneous tumor that rarely recurs after complete excision.

As with pilomatrixoma, pilomatrix carcinomas are asymptomatic and present as solitary dermal or subcutaneous masses3,4 that most commonly are found in the posterior neck, upper back, and preauricular regions of middle-aged or elderly adults with male predominance.5 They range in size from 0.5 to 20 cm with a mean of 4 cm that is slightly larger than pilomatrixoma. Pilomatrix carcinomas predominantly are firm tumors with or without cystic components, and they exhibit a high probability of recurrence and have risk for distant metastasis.6-15

 

 

The differential diagnosis includes epidermal cysts, pilomatrixoma, basal cell carcinoma with matrical differentiation, trichoblastoma/trichoblastic carcinoma, and trichilemmal carcinoma. Pilomatrix carcinomas frequently are mistaken for epidermal cysts on clinical examination. Such a distinction can be easily resolved by histopathologic evaluation. The more challenging differential diagnosis is with pilomatrixoma. Histologically, pilomatrixomas consist of a distinct population of cells including basaloid, squamoid, transitional, and shadow cells in variable proportions. The basaloid cells transition to shadow cells in an organized zonal fashion.16 Compared to pilomatrixomas, pilomatrix carcinomas often show predominance of the basaloid cells; marked cytologic atypia and pleomorphism; numerous mitotic figures; deep infiltrative pattern into subcutaneous fat, fascia, and skeletal muscle; stromal desmoplasia; necrosis; and neurovascular invasion (Tables 1 and 2). Furthermore, the shadow cells tend to form a small nested pattern in pilomatrix carcinoma instead of the flat sheetlike pattern usually observed in pilomatrixoma.16 Basal cell carcinoma with matrical differentiation can pose a diagnostic challenge in the differential diagnosis; basal cell carcinoma usually exhibits a peripheral palisade of the basaloid cells accompanied by retraction spaces separating the tumor from the stroma. Trichoblastoma/trichoblastic carcinoma with matrical differentiation can be distinguished by its exuberant stroma, prominent primitive hair follicles, and papillary mesenchymal bodies. Trichilemmal carcinomas are recognized by their connection to the overlying epidermis, peripheral palisading, and presence of clear cells, while pilomatrix carcinoma lacks connection to the surface epithelium.

Immunohistochemical stains have little to no role in the differential diagnosis, and morphology is the mainstay in making the diagnosis. Rarely, pilomatrix carcinoma can be confused with poorly differentiated sebaceous carcinoma and poorly differentiated squamous cell carcinoma. Although careful scrutiny of the histologic features may help identify mature sebocytes in sebaceous carcinoma, evidence of keratinization in squamous cell carcinoma and ghost cells in pilomatrix carcinoma, using a panel of immunohistochemical stains can be helpful in reaching the final diagnosis (Table 3).

The development of hair matrix tumors have been known to harbor mutations in exon 3 of the catenin beta-1 gene, CTNNB1, that encodes for β-catenin, a downstream effector in the Wnt signaling pathway responsible for differentiation, proliferation, and adhesion of epithelial stem cells.17-21 In a study conducted by Kazakov et al,22 DNA was extracted from 86 lesions: 4 were pilomatrixomas and 1 was a pilomatrix carcinoma. A polymerase chain reaction assay revealed 8 pathogenic variants of the β-catenin gene. D32Y (CTNNB1):c.94G>T (p.Asp32Tyr) and G34R (CTNNB1):c.100G>C (p.Gly34Arg) were the mutations present in pilomatrixoma and pilomatrix carcinoma, respectively.22 In addition, there are several proteins that are part of the Wnt pathway in addition to β-catenin—LEF-1 and CDX-2.

Tumminello and Hosler23 found that pilomatrixomas and pilomatrix carcinomas were positive for CDX-2, β-catenin, and LEF-1 by immunohistochemistry. These downstream molecules in the Wnt signaling pathway could have the potential to be used as diagnostic and prognostic markers.2,13,15,23

Although the pathogenesis is unclear, there are 2 possible mechanisms by which pilomatrix carcinomas develop. They can either arise as de novo tumors, or it is possible that initial mutations in β-catenin result in the formation of pilomatrixomas at an early age that may undergo malignant transformation in elderly patients over time with additional mutations.2

 

 

Our case was strongly and diffusely positive for β-catenin in a nuclear and cytoplasmic pattern and CDX-2 in a nuclear pattern, supporting the role of the Wnt signaling pathway in such tumors. Furthermore, our case demonstrated the presence of few intralesional normal dendritic melanocytes, a rare finding1,24,25 but not unexpected, as melanocytes normally are present within the hair follicle matrix.

Pilomatrix carcinomas are aggressive tumors with a high risk for local recurrence and tendency for metastasis. In a study of 13 cases of pilomatrix carcinomas, Herrmann et al13 found that metastasis was significantly associated with local tumor recurrence (P<.0413). They concluded that the combination of overall high local recurrence and metastatic rates of pilomatrix carcinoma as well as documented tumor-related deaths would warrant continued patient follow-up, especially for recurrent tumors.13 Rapid growth of a tumor, either de novo or following several months of stable size, should alert physicians to perform a diagnostic biopsy.

Management options of pilomatrix carcinoma include surgery or radiation with close follow-up. The most widely reported treatment of pilomatrix carcinoma is wide local excision with histologically confirmed clear margins. Mohs micrographic surgery is an excellent treatment option.2,13-15 Adjuvant radiation therapy may be necessary following excision. Currently there is no consensus on surgical management, and standard excisional margins have not been defined.26 Jones et al2 concluded that complete excision with wide margins likely is curative, with decreased rates of recurrence, and better awareness of this carcinoma would lead to appropriate treatment while avoiding unnecessary diagnostic tests.2

 

Conclusion

We report an exceptionally unique case of early pilomatrix carcinoma with a discussion on the pathogenesis and molecular pathology of hair matrix tumors. A large cohort of patients with longer follow-up periods and better molecular characterization is essential in drawing accurate information about their prognosis, identifying molecular markers that can be used as therapeutic targets, and determining ideal management strategy.

References
  1. Jani P, Chetty R, Ghazarian DM. An unusual composite pilomatrix carcinoma with intralesional melanocytes: differential diagnosis, immunohistochemical evaluation, and review of the literature. Am J Dermatopathol. 2008;30:174-177.
  2. Jones C, Twoon M, Ho W, et al. Pilomatrix carcinoma: 12-year experience and review of the literature. J Cutan Pathol. 2018;45:33-38.
  3. Forbis R, Helwig EB. Pilomatrixoma (calcifying epithelioma). Arch Dermatol. 1961;83:606.
  4. Elder D, Elenitsas R, Ragsdale BD. Tumors of epidermal appendages. In: Elder D, Elenitsas R, Jaworsky C, eds. Lever’s Histopathology of the Skin. 8th ed. Lippincott Raven; 1997:757-759.
  5. Aherne NJ, Fitzpatrick DA, Gibbons D, et al. Pilomatrix carcinoma presenting as an extra axial mass: clinicopathological features. Diagn Pathol. 2008;3:47.
  6. Papadakis M, de Bree E, Floros N, et al. Pilomatrix carcinoma: more malignant biological behavior than was considered in the past. Mol Clin Oncol. 2017;6:415-418.
  7. LeBoit PE, Parslow TG, Choy SH. Hair matrix differentiation: occurrence in lesions other than pilomatricoma. Am J Dermatopathol. 1987;9:399-405.
  8. Campoy F, Stiefel P, Stiefel E, et al. Pilomatrix carcinoma: role played by MR imaging. Neuroradiology. 1989;31:196-198.
  9. Tateyama H, Eimoto T, Tada T, et al. Malignant pilomatricoma: an immunohistochemical study with antihair keratin antibody. Cancer. 1992;69:127-132.
  10. O’Donovan DG, Freemont AJ, Adams JE, et al. Malignant pilomatrixoma with bone metastasis. Histopathology. 1993;23:385-386.
  11. Cross P, Richmond I, Wells S, et al. Malignant pilomatrixoma with bone metastasis. Histopathology. 1994;24:499-500.
  12. Niedermeyer HP, Peris K, Höfler H. Pilomatrix carcinoma with multiple visceral metastases: report of a case. Cancer. 1996;77:1311-1314.
  13. Herrmann JL, Allan A, Trapp KM, et al. Pilomatrix carcinoma: 13 new cases and review of the literature with emphasis on predictors of metastasis. J Am Acad Dermatol. 2014;71:38-43.
  14. Xing L, Marzolf SA, Vandergriff T, et al. Facial pilomatrix carcinomas treated with Mohs micrographic surgery. JAAD Case Rep. 2018;4:253-255.
  15. Fernandez-Flores A, Cassarino DS. Sarcomatoid pilomatrix carcinoma. J Cutan Pathol. 2018;45:508-514.
  16. Sau P, Lupton GP, Graham JH. Pilomatrix carcinoma. Cancer. 1993;71:2491-2498.
  17. Chan E, Gat U, McNiff JM, et al. A common human skin tumour is caused by activating mutations in β-catenin. Nat Genet. 1999;21:410-413.
  18. Huelsken J, Vogel R, Erdmann B, et al. β-catenin controls hair follicle morphogenesis and stem cell differentiation in the skin. Cell. 2001;105:533-545.
  19. Kikuchi A. Tumor formation by genetic mutations in the components of the Wnt signaling pathway. Cancer Sci. 2003;94:225-229.
  20. Durand M, Moles J. Beta-catenin mutations in a common skin cancer: pilomatricoma. Bull Cancer. 1999;86:725-726.
  21. Lazar AJF, Calonje E, Grayson W, et al. Pilomatrix carcinomas contain mutations in CTNNB1, the gene encoding beta-catenin. J Cutan Pathol. 2005;32:148-157.
  22. Kazakov DV, Sima R, Vanecek T, et al. Mutations in exon 3 of the CTNNB1 gene (β-catenin gene) in cutaneous adnexal tumors. Am J Dermatopathol. 2009;31:248-255.
  23. Tumminello K, Hosler GA. CDX2 and LEF-1 expression in pilomatrical tumors and their utility in the diagnosis of pilomatrical carcinoma. J Cutan Pathol. 2018;45:318-324.
  24. Rodic´ N, Taube JM, Manson P, et al Locally invasive dermal squamomelanocytic tumor with matrical differentiation: a peculiar case with review of the literature. Am J Dermatopathol. 2013;35:E72-E76.
  25. Perez C, Debbaneh M, Cassarino D. Preference for the term pilomatrical carcinoma with melanocytic hyperplasia: letter to the editor. J Cutan Pathol. 2017;44:655-657.
  26. Herrmann JL, Allan A, Trapp KM, et al. Pilomatrix carcinoma: 13 new cases and review of the literature with emphasis on predictors of metastasis. J Am Acad Dermatol. 2014;71:38-43.
References
  1. Jani P, Chetty R, Ghazarian DM. An unusual composite pilomatrix carcinoma with intralesional melanocytes: differential diagnosis, immunohistochemical evaluation, and review of the literature. Am J Dermatopathol. 2008;30:174-177.
  2. Jones C, Twoon M, Ho W, et al. Pilomatrix carcinoma: 12-year experience and review of the literature. J Cutan Pathol. 2018;45:33-38.
  3. Forbis R, Helwig EB. Pilomatrixoma (calcifying epithelioma). Arch Dermatol. 1961;83:606.
  4. Elder D, Elenitsas R, Ragsdale BD. Tumors of epidermal appendages. In: Elder D, Elenitsas R, Jaworsky C, eds. Lever’s Histopathology of the Skin. 8th ed. Lippincott Raven; 1997:757-759.
  5. Aherne NJ, Fitzpatrick DA, Gibbons D, et al. Pilomatrix carcinoma presenting as an extra axial mass: clinicopathological features. Diagn Pathol. 2008;3:47.
  6. Papadakis M, de Bree E, Floros N, et al. Pilomatrix carcinoma: more malignant biological behavior than was considered in the past. Mol Clin Oncol. 2017;6:415-418.
  7. LeBoit PE, Parslow TG, Choy SH. Hair matrix differentiation: occurrence in lesions other than pilomatricoma. Am J Dermatopathol. 1987;9:399-405.
  8. Campoy F, Stiefel P, Stiefel E, et al. Pilomatrix carcinoma: role played by MR imaging. Neuroradiology. 1989;31:196-198.
  9. Tateyama H, Eimoto T, Tada T, et al. Malignant pilomatricoma: an immunohistochemical study with antihair keratin antibody. Cancer. 1992;69:127-132.
  10. O’Donovan DG, Freemont AJ, Adams JE, et al. Malignant pilomatrixoma with bone metastasis. Histopathology. 1993;23:385-386.
  11. Cross P, Richmond I, Wells S, et al. Malignant pilomatrixoma with bone metastasis. Histopathology. 1994;24:499-500.
  12. Niedermeyer HP, Peris K, Höfler H. Pilomatrix carcinoma with multiple visceral metastases: report of a case. Cancer. 1996;77:1311-1314.
  13. Herrmann JL, Allan A, Trapp KM, et al. Pilomatrix carcinoma: 13 new cases and review of the literature with emphasis on predictors of metastasis. J Am Acad Dermatol. 2014;71:38-43.
  14. Xing L, Marzolf SA, Vandergriff T, et al. Facial pilomatrix carcinomas treated with Mohs micrographic surgery. JAAD Case Rep. 2018;4:253-255.
  15. Fernandez-Flores A, Cassarino DS. Sarcomatoid pilomatrix carcinoma. J Cutan Pathol. 2018;45:508-514.
  16. Sau P, Lupton GP, Graham JH. Pilomatrix carcinoma. Cancer. 1993;71:2491-2498.
  17. Chan E, Gat U, McNiff JM, et al. A common human skin tumour is caused by activating mutations in β-catenin. Nat Genet. 1999;21:410-413.
  18. Huelsken J, Vogel R, Erdmann B, et al. β-catenin controls hair follicle morphogenesis and stem cell differentiation in the skin. Cell. 2001;105:533-545.
  19. Kikuchi A. Tumor formation by genetic mutations in the components of the Wnt signaling pathway. Cancer Sci. 2003;94:225-229.
  20. Durand M, Moles J. Beta-catenin mutations in a common skin cancer: pilomatricoma. Bull Cancer. 1999;86:725-726.
  21. Lazar AJF, Calonje E, Grayson W, et al. Pilomatrix carcinomas contain mutations in CTNNB1, the gene encoding beta-catenin. J Cutan Pathol. 2005;32:148-157.
  22. Kazakov DV, Sima R, Vanecek T, et al. Mutations in exon 3 of the CTNNB1 gene (β-catenin gene) in cutaneous adnexal tumors. Am J Dermatopathol. 2009;31:248-255.
  23. Tumminello K, Hosler GA. CDX2 and LEF-1 expression in pilomatrical tumors and their utility in the diagnosis of pilomatrical carcinoma. J Cutan Pathol. 2018;45:318-324.
  24. Rodic´ N, Taube JM, Manson P, et al Locally invasive dermal squamomelanocytic tumor with matrical differentiation: a peculiar case with review of the literature. Am J Dermatopathol. 2013;35:E72-E76.
  25. Perez C, Debbaneh M, Cassarino D. Preference for the term pilomatrical carcinoma with melanocytic hyperplasia: letter to the editor. J Cutan Pathol. 2017;44:655-657.
  26. Herrmann JL, Allan A, Trapp KM, et al. Pilomatrix carcinoma: 13 new cases and review of the literature with emphasis on predictors of metastasis. J Am Acad Dermatol. 2014;71:38-43.
Issue
Cutis - 108(4)
Issue
Cutis - 108(4)
Page Number
E24-E28
Page Number
E24-E28
Publications
Publications
Topics
Article Type
Display Headline
Early Pilomatrix Carcinoma: A Case Report With Emphasis on Molecular Pathology and Review of the Literature
Display Headline
Early Pilomatrix Carcinoma: A Case Report With Emphasis on Molecular Pathology and Review of the Literature
Sections
Inside the Article

Practice Points

  • Clinicians and pathologists should be aware of pilomatrix carcinoma to facilitate early detection.
  • Early diagnosis and prompt treatment of pilomatrix carcinoma is crucial in lowering recurrence rate and avoiding a poor outcome.
  • Caudal-related homeobox transcription factor 2 and β-catenin components of the Wnt signaling pathway play an important role in the pathogenesis of pilomatrix carcinoma.
  • Although controversial, wide local excision is the treatment of choice for pilomatrix carcinoma.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

75-year-old woman • right-side rib pain • radiating shoulder pain • history of hypertension & hypercholesterolemia • Dx?

Article Type
Changed
Wed, 11/17/2021 - 08:23
Display Headline
75-year-old woman • right-side rib pain • radiating shoulder pain • history of hypertension & hypercholesterolemia • Dx?

THE CASE

A 75-year-old woman presented to the primary care clinic with right-side rib pain. The patient said the pain started 1 week earlier, after she ate fried chicken for dinner, and had since been exacerbated by rich meals, lying supine, and taking a deep inspiratory breath. She also said that prior to coming to the clinic that day, the pain had been radiating to her right shoulder.

The patient denied experiencing associated fevers, chills, shortness of breath, chest pain, nausea, vomiting, constipation, diarrhea, or changes in stool color. She had a history of hypertension, for which she was taking lisinopril 20 mg/d, and hypercholesterolemia, for which she was on simvastatin 10 mg/d. She was additionally using timolol ophthalmic solution for her glaucoma.

During the examination, the patient’s vital signs were stable, with a pulse of 80 beats/min, a respiratory rate of 16 breaths/min, and an oxygen saturation of 98% on room air. The patient had no abdominal tenderness upon palpation, and the physical exam revealed no abnormalities. An in-office electrocardiogram was performed, with normal results. Additionally, a comprehensive metabolic panel, lipase test, and d-dimer test were ordered. Lab results showed an isolated elevated d-dimer of 2.66 mcg/mL (normal range, < 0.54 mcg/mL), while all other labs were normal.

THE DIAGNOSIS

Based on the lab results, a stat computed tomography pulmonary angiogram (CTPA) was ordered and showed a right segmental and subsegmental pulmonary embolism (PE; FIGURE 1).

A computed tomography pulmonary angiogram

DISCUSSION

PE shares pathophysiologic mechanisms with deep vein thrombosis (DVT), and together these comprise venous thromboembolism (VTE). Risk factors for VTE include hypercoagulable disorders, use of estrogens, active malignancy, and immobilization.1 Unprovoked VTE occurs in the absence of identifiable risk factors and carries a higher risk of recurrence.2,3 While PE is classically thought to occur in the setting of a DVT, there is increasing literature describing de novo PE that can occur independent of a DVT.4

Common symptoms of PE include tachycardia, tachypnea, and pleuritic chest pain.5 Abdominal pain is a rare symptom described in some case reports.6,7 Thus, a high clinical suspicion is needed for diagnosis of PE.

The Wells criteria is an established model for risk stratifying patients presenting with possible VTE (TABLE).8 For patients with low pretest probability, as in this case, a d-dimer is an effective diagnostic work-up, as a negative result will rule out PE. (If the d-dimer had been negative in this case, we would have considered other diagnoses, such as acute coronary syndrome, biliary colic, gastritis, pancreatitis, or musculoskeletal pain.) For high-risk patients, immediate anticoagulation and imaging should be performed, frequently with heparin and CTPA.9

Wells criteria for venous thromboembolism

Continue to: Length of treatment depends on gender and etiology

 

 

Length of treatment depends on gender and etiology

The cornerstone treatment for stable patients with VTE is therapeutic anticoagulation. The new oral anticoagulants, which directly inhibit factor Xa or thrombin, have become increasingly popular for management of VTE, in part because they don’t require INR testing and monitoring.2

The duration of anticoagulation, particularly in unprovoked PE, is debatable. As noted earlier, patients with an unprovoked PE are at higher risk of recurrence than those with a reversible cause, so the question becomes whether these patients should have indefinite anticoagulation.2,3 Studies examining risk stratification of patients with a first, unprovoked VTE have found that men have the highest risk of recurrence, followed by women who were not taking estrogen during the index VTE, and lastly women who were taking estrogen therapy during the index VTE and subsequently discontinued it.2,3,10

Thus, it is reasonable to give women the option to discontinue anticoagulation in the setting of a negative d-dimer follow-up.3 The 2016 CHEST guidelines recommend extended anticoagulation for a first-time, unprovoked VTE, but acknowledge this recommendation is strongest for men and that women with negative d-dimer assays may consider discontinuation.10

Our patient was directed to the emergency department for further monitoring following CT confirmation. She was discharged home after being deemed stable and prescribed apixaban 10 mg/d. A venous duplex ultrasound performed 12 days later for knee pain revealed no venous thrombosis. A CT of the abdomen performed 3 months later for other reasons revealed a normal gallbladder with no visible stones.

Apixaban was continued for 3 months and discontinued after discussion of risks and benefits of therapy cessation in the setting of a normal d-dimer and the 2016 CHEST guidelines for anticoagulation in VTE.10

Continue to: THE TAKEAWAY

 

 

THE TAKEAWAY

PE carries a significantly high mortality rate and can manifest with nonspecific and masquerading signs. A high index of suspicion is required to place PE on the differential diagnosis and carry out appropriate testing. Our patient presented with a history consistent with biliary colic but with pleuritic chest pain that warranted consideration of a PE.

CORRESPONDENCE
Alyssa Anderson, MD, 1 Continental Drive, Elizabethtown, PA 17022; [email protected]

References

1. Israel HL, Goldstein F. The varied clinical manifestations of pulmonary embolism. Ann Intern Med. 1957;47:202-226. doi: 10.7326/0003-4819-47-2-202

2. Rehman H, John E, Parikh P. Pulmonary embolism presenting as abdominal pain: an atypical presentation of a common diagnosis. Case Rep Emerg Med. 2016;2016:1-3. doi: 10.1155/2016/7832895

3. Park ES, Cho JY, Seo J-H, et al. Pulmonary embolism presenting with acute abdominal pain in a girl with stable ankle fracture and inherited antithrombin deficiency. Blood Res. 2018;53:81-83. doi: 10.5045/br.2018.53.1.81

4. Tapson VF. Acute pulmonary embolism. N Engl J Med. 2008;358:1037-1052. doi: 10.1056/NEJMra072753

5. Agrawal V, Kim ESH. Risk of recurrent venous thromboembolism after an initial episode: risk stratification and implications for long-term treatment. Curr Cardiol Rep. 2019;21:24. doi: 10.1007/s11886-019-1111-2

6. Kearon C, Parpia S, Spencer FA, et al. Long‐term risk of recurrence in patients with a first unprovoked venous thromboembolism managed according to d‐dimer results; A cohort study. J Thromb Haemost. 2019;17:1144-1152. doi: 10.1111/jth.14458

7. Van Gent J-M, Zander AL, Olson EJ, et al. Pulmonary embolism without deep venous thrombosis. J Trauma Acute Care Surg. 2014;76:1270-1274. doi: 10.1097/TA.0000000000000233

8. Wells PS, Anderson DR, Rodger M, et al. Excluding pulmonary embolism at the bedside without diagnostic imaging: management of patients with suspected pulmonary embolism presenting to the emergency department by using a simple clinical model and d-dimer. Ann Intern Med. 2001;135:98-107. doi: 10.7326/0003-4819-135-2-200107170-00010

9. Kline JA. Diagnosis and exclusion of pulmonary embolism. Thromb Res. 2018;163:207-220. doi: 10.1016/j.thromres.2017.06.002

10. Kearon C, Akl EA, Ornelas J, et al. Antithrombotic therapy for VTE disease. Chest. 2016;149:315-352. doi: 10.1016/j.chest.2015.11.026

Article PDF
Author and Disclosure Information

Penn State College of Medicine, Penn State University, Hershey (Dr. Albertson); Family & Community Medicine, Penn State Health Milton S. Hershey Medical Center, Hershey (Dr. Anderson)
[email protected]

The authors reported no potential conflict of interest relevant to this article.

Issue
The Journal of Family Practice - 70(8)
Publications
Topics
Page Number
405-407
Sections
Author and Disclosure Information

Penn State College of Medicine, Penn State University, Hershey (Dr. Albertson); Family & Community Medicine, Penn State Health Milton S. Hershey Medical Center, Hershey (Dr. Anderson)
[email protected]

The authors reported no potential conflict of interest relevant to this article.

Author and Disclosure Information

Penn State College of Medicine, Penn State University, Hershey (Dr. Albertson); Family & Community Medicine, Penn State Health Milton S. Hershey Medical Center, Hershey (Dr. Anderson)
[email protected]

The authors reported no potential conflict of interest relevant to this article.

Article PDF
Article PDF

THE CASE

A 75-year-old woman presented to the primary care clinic with right-side rib pain. The patient said the pain started 1 week earlier, after she ate fried chicken for dinner, and had since been exacerbated by rich meals, lying supine, and taking a deep inspiratory breath. She also said that prior to coming to the clinic that day, the pain had been radiating to her right shoulder.

The patient denied experiencing associated fevers, chills, shortness of breath, chest pain, nausea, vomiting, constipation, diarrhea, or changes in stool color. She had a history of hypertension, for which she was taking lisinopril 20 mg/d, and hypercholesterolemia, for which she was on simvastatin 10 mg/d. She was additionally using timolol ophthalmic solution for her glaucoma.

During the examination, the patient’s vital signs were stable, with a pulse of 80 beats/min, a respiratory rate of 16 breaths/min, and an oxygen saturation of 98% on room air. The patient had no abdominal tenderness upon palpation, and the physical exam revealed no abnormalities. An in-office electrocardiogram was performed, with normal results. Additionally, a comprehensive metabolic panel, lipase test, and d-dimer test were ordered. Lab results showed an isolated elevated d-dimer of 2.66 mcg/mL (normal range, < 0.54 mcg/mL), while all other labs were normal.

THE DIAGNOSIS

Based on the lab results, a stat computed tomography pulmonary angiogram (CTPA) was ordered and showed a right segmental and subsegmental pulmonary embolism (PE; FIGURE 1).

A computed tomography pulmonary angiogram

DISCUSSION

PE shares pathophysiologic mechanisms with deep vein thrombosis (DVT), and together these comprise venous thromboembolism (VTE). Risk factors for VTE include hypercoagulable disorders, use of estrogens, active malignancy, and immobilization.1 Unprovoked VTE occurs in the absence of identifiable risk factors and carries a higher risk of recurrence.2,3 While PE is classically thought to occur in the setting of a DVT, there is increasing literature describing de novo PE that can occur independent of a DVT.4

Common symptoms of PE include tachycardia, tachypnea, and pleuritic chest pain.5 Abdominal pain is a rare symptom described in some case reports.6,7 Thus, a high clinical suspicion is needed for diagnosis of PE.

The Wells criteria is an established model for risk stratifying patients presenting with possible VTE (TABLE).8 For patients with low pretest probability, as in this case, a d-dimer is an effective diagnostic work-up, as a negative result will rule out PE. (If the d-dimer had been negative in this case, we would have considered other diagnoses, such as acute coronary syndrome, biliary colic, gastritis, pancreatitis, or musculoskeletal pain.) For high-risk patients, immediate anticoagulation and imaging should be performed, frequently with heparin and CTPA.9

Wells criteria for venous thromboembolism

Continue to: Length of treatment depends on gender and etiology

 

 

Length of treatment depends on gender and etiology

The cornerstone treatment for stable patients with VTE is therapeutic anticoagulation. The new oral anticoagulants, which directly inhibit factor Xa or thrombin, have become increasingly popular for management of VTE, in part because they don’t require INR testing and monitoring.2

The duration of anticoagulation, particularly in unprovoked PE, is debatable. As noted earlier, patients with an unprovoked PE are at higher risk of recurrence than those with a reversible cause, so the question becomes whether these patients should have indefinite anticoagulation.2,3 Studies examining risk stratification of patients with a first, unprovoked VTE have found that men have the highest risk of recurrence, followed by women who were not taking estrogen during the index VTE, and lastly women who were taking estrogen therapy during the index VTE and subsequently discontinued it.2,3,10

Thus, it is reasonable to give women the option to discontinue anticoagulation in the setting of a negative d-dimer follow-up.3 The 2016 CHEST guidelines recommend extended anticoagulation for a first-time, unprovoked VTE, but acknowledge this recommendation is strongest for men and that women with negative d-dimer assays may consider discontinuation.10

Our patient was directed to the emergency department for further monitoring following CT confirmation. She was discharged home after being deemed stable and prescribed apixaban 10 mg/d. A venous duplex ultrasound performed 12 days later for knee pain revealed no venous thrombosis. A CT of the abdomen performed 3 months later for other reasons revealed a normal gallbladder with no visible stones.

Apixaban was continued for 3 months and discontinued after discussion of risks and benefits of therapy cessation in the setting of a normal d-dimer and the 2016 CHEST guidelines for anticoagulation in VTE.10

Continue to: THE TAKEAWAY

 

 

THE TAKEAWAY

PE carries a significantly high mortality rate and can manifest with nonspecific and masquerading signs. A high index of suspicion is required to place PE on the differential diagnosis and carry out appropriate testing. Our patient presented with a history consistent with biliary colic but with pleuritic chest pain that warranted consideration of a PE.

CORRESPONDENCE
Alyssa Anderson, MD, 1 Continental Drive, Elizabethtown, PA 17022; [email protected]

THE CASE

A 75-year-old woman presented to the primary care clinic with right-side rib pain. The patient said the pain started 1 week earlier, after she ate fried chicken for dinner, and had since been exacerbated by rich meals, lying supine, and taking a deep inspiratory breath. She also said that prior to coming to the clinic that day, the pain had been radiating to her right shoulder.

The patient denied experiencing associated fevers, chills, shortness of breath, chest pain, nausea, vomiting, constipation, diarrhea, or changes in stool color. She had a history of hypertension, for which she was taking lisinopril 20 mg/d, and hypercholesterolemia, for which she was on simvastatin 10 mg/d. She was additionally using timolol ophthalmic solution for her glaucoma.

During the examination, the patient’s vital signs were stable, with a pulse of 80 beats/min, a respiratory rate of 16 breaths/min, and an oxygen saturation of 98% on room air. The patient had no abdominal tenderness upon palpation, and the physical exam revealed no abnormalities. An in-office electrocardiogram was performed, with normal results. Additionally, a comprehensive metabolic panel, lipase test, and d-dimer test were ordered. Lab results showed an isolated elevated d-dimer of 2.66 mcg/mL (normal range, < 0.54 mcg/mL), while all other labs were normal.

THE DIAGNOSIS

Based on the lab results, a stat computed tomography pulmonary angiogram (CTPA) was ordered and showed a right segmental and subsegmental pulmonary embolism (PE; FIGURE 1).

A computed tomography pulmonary angiogram

DISCUSSION

PE shares pathophysiologic mechanisms with deep vein thrombosis (DVT), and together these comprise venous thromboembolism (VTE). Risk factors for VTE include hypercoagulable disorders, use of estrogens, active malignancy, and immobilization.1 Unprovoked VTE occurs in the absence of identifiable risk factors and carries a higher risk of recurrence.2,3 While PE is classically thought to occur in the setting of a DVT, there is increasing literature describing de novo PE that can occur independent of a DVT.4

Common symptoms of PE include tachycardia, tachypnea, and pleuritic chest pain.5 Abdominal pain is a rare symptom described in some case reports.6,7 Thus, a high clinical suspicion is needed for diagnosis of PE.

The Wells criteria is an established model for risk stratifying patients presenting with possible VTE (TABLE).8 For patients with low pretest probability, as in this case, a d-dimer is an effective diagnostic work-up, as a negative result will rule out PE. (If the d-dimer had been negative in this case, we would have considered other diagnoses, such as acute coronary syndrome, biliary colic, gastritis, pancreatitis, or musculoskeletal pain.) For high-risk patients, immediate anticoagulation and imaging should be performed, frequently with heparin and CTPA.9

Wells criteria for venous thromboembolism

Continue to: Length of treatment depends on gender and etiology

 

 

Length of treatment depends on gender and etiology

The cornerstone treatment for stable patients with VTE is therapeutic anticoagulation. The new oral anticoagulants, which directly inhibit factor Xa or thrombin, have become increasingly popular for management of VTE, in part because they don’t require INR testing and monitoring.2

The duration of anticoagulation, particularly in unprovoked PE, is debatable. As noted earlier, patients with an unprovoked PE are at higher risk of recurrence than those with a reversible cause, so the question becomes whether these patients should have indefinite anticoagulation.2,3 Studies examining risk stratification of patients with a first, unprovoked VTE have found that men have the highest risk of recurrence, followed by women who were not taking estrogen during the index VTE, and lastly women who were taking estrogen therapy during the index VTE and subsequently discontinued it.2,3,10

Thus, it is reasonable to give women the option to discontinue anticoagulation in the setting of a negative d-dimer follow-up.3 The 2016 CHEST guidelines recommend extended anticoagulation for a first-time, unprovoked VTE, but acknowledge this recommendation is strongest for men and that women with negative d-dimer assays may consider discontinuation.10

Our patient was directed to the emergency department for further monitoring following CT confirmation. She was discharged home after being deemed stable and prescribed apixaban 10 mg/d. A venous duplex ultrasound performed 12 days later for knee pain revealed no venous thrombosis. A CT of the abdomen performed 3 months later for other reasons revealed a normal gallbladder with no visible stones.

Apixaban was continued for 3 months and discontinued after discussion of risks and benefits of therapy cessation in the setting of a normal d-dimer and the 2016 CHEST guidelines for anticoagulation in VTE.10

Continue to: THE TAKEAWAY

 

 

THE TAKEAWAY

PE carries a significantly high mortality rate and can manifest with nonspecific and masquerading signs. A high index of suspicion is required to place PE on the differential diagnosis and carry out appropriate testing. Our patient presented with a history consistent with biliary colic but with pleuritic chest pain that warranted consideration of a PE.

CORRESPONDENCE
Alyssa Anderson, MD, 1 Continental Drive, Elizabethtown, PA 17022; [email protected]

References

1. Israel HL, Goldstein F. The varied clinical manifestations of pulmonary embolism. Ann Intern Med. 1957;47:202-226. doi: 10.7326/0003-4819-47-2-202

2. Rehman H, John E, Parikh P. Pulmonary embolism presenting as abdominal pain: an atypical presentation of a common diagnosis. Case Rep Emerg Med. 2016;2016:1-3. doi: 10.1155/2016/7832895

3. Park ES, Cho JY, Seo J-H, et al. Pulmonary embolism presenting with acute abdominal pain in a girl with stable ankle fracture and inherited antithrombin deficiency. Blood Res. 2018;53:81-83. doi: 10.5045/br.2018.53.1.81

4. Tapson VF. Acute pulmonary embolism. N Engl J Med. 2008;358:1037-1052. doi: 10.1056/NEJMra072753

5. Agrawal V, Kim ESH. Risk of recurrent venous thromboembolism after an initial episode: risk stratification and implications for long-term treatment. Curr Cardiol Rep. 2019;21:24. doi: 10.1007/s11886-019-1111-2

6. Kearon C, Parpia S, Spencer FA, et al. Long‐term risk of recurrence in patients with a first unprovoked venous thromboembolism managed according to d‐dimer results; A cohort study. J Thromb Haemost. 2019;17:1144-1152. doi: 10.1111/jth.14458

7. Van Gent J-M, Zander AL, Olson EJ, et al. Pulmonary embolism without deep venous thrombosis. J Trauma Acute Care Surg. 2014;76:1270-1274. doi: 10.1097/TA.0000000000000233

8. Wells PS, Anderson DR, Rodger M, et al. Excluding pulmonary embolism at the bedside without diagnostic imaging: management of patients with suspected pulmonary embolism presenting to the emergency department by using a simple clinical model and d-dimer. Ann Intern Med. 2001;135:98-107. doi: 10.7326/0003-4819-135-2-200107170-00010

9. Kline JA. Diagnosis and exclusion of pulmonary embolism. Thromb Res. 2018;163:207-220. doi: 10.1016/j.thromres.2017.06.002

10. Kearon C, Akl EA, Ornelas J, et al. Antithrombotic therapy for VTE disease. Chest. 2016;149:315-352. doi: 10.1016/j.chest.2015.11.026

References

1. Israel HL, Goldstein F. The varied clinical manifestations of pulmonary embolism. Ann Intern Med. 1957;47:202-226. doi: 10.7326/0003-4819-47-2-202

2. Rehman H, John E, Parikh P. Pulmonary embolism presenting as abdominal pain: an atypical presentation of a common diagnosis. Case Rep Emerg Med. 2016;2016:1-3. doi: 10.1155/2016/7832895

3. Park ES, Cho JY, Seo J-H, et al. Pulmonary embolism presenting with acute abdominal pain in a girl with stable ankle fracture and inherited antithrombin deficiency. Blood Res. 2018;53:81-83. doi: 10.5045/br.2018.53.1.81

4. Tapson VF. Acute pulmonary embolism. N Engl J Med. 2008;358:1037-1052. doi: 10.1056/NEJMra072753

5. Agrawal V, Kim ESH. Risk of recurrent venous thromboembolism after an initial episode: risk stratification and implications for long-term treatment. Curr Cardiol Rep. 2019;21:24. doi: 10.1007/s11886-019-1111-2

6. Kearon C, Parpia S, Spencer FA, et al. Long‐term risk of recurrence in patients with a first unprovoked venous thromboembolism managed according to d‐dimer results; A cohort study. J Thromb Haemost. 2019;17:1144-1152. doi: 10.1111/jth.14458

7. Van Gent J-M, Zander AL, Olson EJ, et al. Pulmonary embolism without deep venous thrombosis. J Trauma Acute Care Surg. 2014;76:1270-1274. doi: 10.1097/TA.0000000000000233

8. Wells PS, Anderson DR, Rodger M, et al. Excluding pulmonary embolism at the bedside without diagnostic imaging: management of patients with suspected pulmonary embolism presenting to the emergency department by using a simple clinical model and d-dimer. Ann Intern Med. 2001;135:98-107. doi: 10.7326/0003-4819-135-2-200107170-00010

9. Kline JA. Diagnosis and exclusion of pulmonary embolism. Thromb Res. 2018;163:207-220. doi: 10.1016/j.thromres.2017.06.002

10. Kearon C, Akl EA, Ornelas J, et al. Antithrombotic therapy for VTE disease. Chest. 2016;149:315-352. doi: 10.1016/j.chest.2015.11.026

Issue
The Journal of Family Practice - 70(8)
Issue
The Journal of Family Practice - 70(8)
Page Number
405-407
Page Number
405-407
Publications
Publications
Topics
Article Type
Display Headline
75-year-old woman • right-side rib pain • radiating shoulder pain • history of hypertension & hypercholesterolemia • Dx?
Display Headline
75-year-old woman • right-side rib pain • radiating shoulder pain • history of hypertension & hypercholesterolemia • Dx?
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Acyclovir-Resistant Cutaneous Herpes Simplex Virus in DOCK8 Deficiency  

Article Type
Changed
Thu, 10/14/2021 - 16:10
Display Headline
Acyclovir-Resistant Cutaneous Herpes Simplex Virus in DOCK8 Deficiency  

Dedicator of cytokinesis 8 (DOCK8 ) deficiency is the major cause of autosomal-recessive hyper-IgEsyndrome. 1 Characteristic clinical features including eosinophilia, eczema, and recurrent Staphylococcus aureus cutaneous and respiratory tract infections are common in DOCK8 deficiency, similar to the autosomal-dominant form of hyper-IgE syndrome that is due to defi c iency of signal transducer and activation of transcription 3 (STAT-3 ). 1 In addition, patients with DOCK8 deficiency are particularly susceptible to asthma; food allergies; lymphomas; and severe cutaneous viral infections, including herpes simplex virus (HSV), molluscum contagiosum, varicella-zoster virus, and human papillomavirus. Since the discovery of the DOCK8 gene in 2009, various studies have sought to elucidate the mechanistic contribution of DOCK8 to the dermatologic immune environment. 2 Although cutaneous viral infections such as those caused by HSV typically are short lived and self-limiting in immunocompetent hosts, they have proven to be severe and recalcitrant in the setting of DOCK8 deficiency. 1 Herein, we report the case of a 32-month-old girl with homozygous DOCK8 deficiency who developed acyclovir-resistant cutaneous HSV. 

Case Report

A 32-month-old girl presented with an approximately 2-cm linear erosion along the left posterior auricular sulcus at month 9 of a hospital stay for recurrent infections. Her medical history was notable for multiple upper respiratory tract infections, diffuse eczema, and food allergies. She had presented to an outside hospital at 14 months of age with herpetic gingivostomatitis and eczema herpeticum that was successfully treated with acyclovir. She was readmitted at 20 months of age due to Pneumocystis jiroveci pneumonia, pancytopenia, and disseminated histoplasmosis. Prophylactic oral acyclovir (20 mg/kg twice daily) was started, given her history of HSV infection. Because of recurrent infections, she underwent an immunodeficiency workup. Whole exome sequencing analysis revealed a homozygous deletion c.(528+1_5291)_(1516+1_15171)del in DOCK8 gene–affecting exons 5 to 13. The patient was transferred to our hospital for continued care and as a potential candidate for bone marrow transplant following resolution of the disseminated histoplasmosis infection.

During her hospitalization at the current presentation, she was noted to have a 2-cm linear erosion along the left posterior auricular sulcus. Initial wound care with bacitracin ointment was applied to the area while specimens were obtained and empiric oral acyclovir therapy was initiated (20 mg/kg 4 times daily [QID]), given a clinical impression consistent with cutaneous HSV infection despite acyclovir prophylaxis. Direct immunofluorescence and viral cultures were positive for HSV-1, while bacterial cultures grew methicillin-susceptible S aureus. Cephalexin and mupirocin ointment were started, and acyclovir was continued. After 2 weeks of therapy, there was no visible change in the wound; cultures were repeated, again showing the wound contained HSV. Bacterial cultures this time grew Pseudomonas putida, and the antibiotic regimen was transitioned to cefepime.

After no response to the continued course of therapeutic acyclovir, HSV cultures were sent to the Centers for Disease Control and Prevention for resistance testing, and biopsy of the lesion was performed by the otolaryngology service to rule out malignancy and potential alternative diagnoses. Histopathology showed only reactive inflammation without visible microorganisms on tissue HSV-1/HSV-2 immunostain; however, tissue viral culture was positive for HSV-1. The patient was transitioned back to acyclovir (intravenous [IV] 20 mg/kg QID) with the addition of empiric foscarnet (IV 40 mg/kg 3 times daily) given the worsening appearance of the lesion.  The HSV acyclovir resistance test results from the Centers for Disease Control and Prevention returned soon after and were positive for resistance (median infectious dose, 3.29 µg/L [reference interval, sensitive <2.00 µg/L; resistant >1.90 µg/L]). The patient completed a 21-day course of combination foscarnet and acyclovir therapy, during which time the lesion showed notable improvement and healing. The patient was continued on prophylactic acyclovir (IV 20 mg/kg QID). Unfortunately, the patient eventually died due to complications related to pneumonia.

Comment

Infection in Patients With DOCK8 Deficiency—The gene DOCK8 has emerged as playing a central role in both innate and adaptive immunity, as it is expressed primarily in immune cells and serves as a mediator of numerous processes, including immune synapse formation, cell signaling and trafficking, antibody and cytokine production, and lymphocyte memory.3 Cells that are critical for combating cutaneous viral infections, including skin-resident memory T cells and natural killer cells, are defective, which leads to a severely immunocompromised state in DOCK8-deficient patients with a particular susceptibility to infectious and inflammatory dermatologic disease.4 

Herpes simplex virus infection commonly is seen in DOCK8 deficiency, with retrospective analysis of a DOCK8-deficient cohort revealing HSV infection in approximately 38% of patients.5 Prophylactic acyclovir is essential for DOCK8-deficient individuals with a history of HSV infection given the tendency of the virus to reactivate.6 However, despite prophylaxis, our patient developed an HSV-positive posterior auricular erosion that continued to progress even after increase of the acyclovir dose. Acyclovir resistance testing of the HSV isolated from the wound was positive, confirming the clinical suspicion of the presence of acyclovir-resistant HSV infection.

Acyclovir-Resistant HSV—Acyclovir-resistant HSV in immunosuppressed individuals was first noted in 1982, and most cases since then have occurred in the setting of AIDS and in organ transplant recipients.6 Few reports of acyclovir-resistant HSV in DOCK8 deficiency exist, and to our knowledge, our patient is the youngest DOCK8-deficient individual to be documented with acyclovir-resistant HSV infection.1,7-15 We identified relevant cases from the PubMed and EMBASE databases using the search terms DOCK8 deficiency and acyclovir and DOCK8 deficiency and herpes. The eTable lists other reported cases of acyclovir-resistant HSV in DOCK8-deficient patients. The majority of cases involved school-aged females. Lesion types varied and included herpes labialis, eczema herpeticum, and blepharoconjunctivitis. Escalation of therapy and resolution of the lesion was seen in some cases with administration of subcutaneous pegylated interferon alfa-2b.

 

 

Treatment Alternatives—Acyclovir competitively inhibits viral DNA polymerase by incorporating into elongating viral DNA strands and halting chain synthesis. Acyclovir requires triphosphorylation for activation, and viral thymidine kinase is responsible for the first phosphorylation event. Ninety-five percent of cases of acyclovir resistance are secondary to mutations in viral thymidine kinase. Foscarnet also inhibits viral DNA polymerase but does so directly without the need to be phosphorylated first.6 For this reason, foscarnet often is the drug of choice in the treatment of acyclovir-resistant HSV, as evidenced in our patient. However, foscarnet-resistant HSV strains may develop from mutations in the DNA polymerase gene.

Cidofovir is a nucleotide analogue that requires phosphorylation by host, as opposed to viral, kinases for antiviral activity. Intravenous and topical formulations of cidofovir have proven effective in the treatment of acyclovir- and foscarnet-resistant HSV lesions.6 Cidofovir also can be applied intralesionally, a method that provides targeted therapy and minimizes cidofovir-associated nephrotoxicity.12 Reports of systemic interferon alfa therapy for acyclovir-resistant HSV also exist. A study found IFN-⍺ production by peripheral blood mononuclear cells in DOCK8-deficient individuals to be significantly reduced relative to controls (P<.05).7 There has been complete resolution of acyclovir-resistant HSV lesions with subcutaneous pegylated interferon alfa-2b injections in several DOCK8-deficient patients.7-9

The need for escalating therapy in DOCK8-deficient individuals with acyclovir-resistant HSV infection underscores the essential role of DOCK8 in dermatologic immunity. Our case demonstrates that a high degree of suspicion for cutaneous HSV infection should be adopted in DOCK8-deficient patients of any age, regardless of acyclovir prophylaxis. Viral culture in addition to bacterial cultures should be performed early in patients with cutaneous erosions, and the threshold for HSV resistance testing should be low to minimize morbidity associated with these infections. Early resistance testing in our case could have prevented prolongation of infection and likely eliminated the need for a biopsy.

Conclusion

DOCK8 deficiency presents a unique challenge to dermatologists and other health care providers given the susceptibility of affected individuals to developing a reservoir of severe and potentially resistant viral cutaneous infections. Prophylactic acyclovir may not be sufficient for HSV suppression, even in the youngest of patients, and suspicion for resistance should be high to avoid delays in adequate treatment.

References
  1. Chu EY, Freeman AF, Jing H, et al. Cutaneous manifestations of DOCK8 deficiency syndrome. Arch Dermatol. 2012;148:79-84. doi:10.1001/archdermatol.2011.262
  2. Aydin SE, Kilic SS, Aytekin C, et al. DOCK8 deficiency: clinical and immunological phenotype and treatment options—a review of 136 patients. J Clin Immunol. 2015;35:189-198. doi:10.1007/s10875-014-0126-0
  3. Kearney CJ, Randall KL, Oliaro J. DOCK8 regulates signal transduction events to control immunity. Cell Mol Immunol. 2017;14:406-411. doi:10.1038/cmi.2017.9
  4. Zhang Q, Dove CG, Hor JL, et al. DOCK8 regulates lymphocyte shape integrity for skin antiviral immunity. J Exp Med. 2014;211:2549-2566. doi:10.1084/jem.20141307
  5. Engelhardt KR, Gertz EM, Keles S, et al. The extended clinical phenotype of 64 patients with DOCK8 deficiency. J Allergy Clin Immunol. 2015;136:402-412. doi:10.1016/j.jaci.2014.12.1945
  6. Chilukuri S, Rosen T. Management of acyclovir-resistant herpes simplex virus. Dermatol Clin. 2003;21:311-320. doi:10.1016/S0733-8635(02)00093-1
  7. Keles S, Jabara HH, Reisli I, et al. Plasmacytoid dendritic cell depletion in DOCK8 deficiency: rescue of severe herpetic infections with interferon alpha-2b therapy. J Allergy Clin Immunol. 2014;133:1753-1755.e3. doi:10.1016/j.jaci.2014.03.032
  8. Papan C, Hagl B, Heinz V, et al Beneficial IFN-α treatment of tumorous herpes simplex blepharoconjunctivitis in dedicator of cytokinesis 8 deficiency. J Allergy Clin Immunol. 2014;133:1456-1458. doi:10.1016/j.jaci.2014.02.008
  9. Metin A, Kanik-Yuksek S, Ozkaya-Parlakay A, et al. Giant herpes labialis in a child with DOCK8-deficient hyper-IgE syndrome. Pediatr Neonatol. 2016;57:79-80. doi:10.1016/j.pedneo.2015.04.011
  10. Zhang Q, Davis JC, Lamborn IT, et al. Combined immunodeficiency associated with DOCK8 mutations. N Engl J Med. 2009;361:2046-2055. doi:10.1056/NEJMoa0905506
  11. Lei JY, Wang Y, Jaffe ES, et al. Microcystic adnexal carcinoma associated with primary immunodeficiency, recurrent diffuse herpes simplex virus infection, and cutaneous T-cell lymphoma. Am J Dermatopathol. 2000;22:524-529. doi:10.1097/00000372-200012000-00008
  12. Castelo-Soccio L, Bernardin R, Stern J, et al. Successful treatment of acyclovir-resistant herpes simplex virus with intralesional cidofovir. Arch Dermatol. 2010;146:124-126. doi:10.1001/archdermatol.2009.363
  13. Shah NN, Freeman AF, Hickstein DD. Addendum to: haploidentical related donor hematopoietic stem cell transplantation for DOCK8 deficiency using post-transplantation cyclophosphamide. Biol Blood Marrow Transplant. 2019;25:E65-E67. doi:10.1016/j.bbmt.2018.11.014
  14. Freeman AF, Yazigi N, Shah NN, et al. Tandem orthotopic living donor liver transplantation followed by same donor haploidentical hematopoietic stem cell transplantation for DOCK8 deficiency. Transplantation. 2019;103:2144-2149. doi:10.1097/TP.0000000000002649
  15. Casto AM, Stout SC, Selvarangan R, et al. Evaluation of genotypic antiviral resistance testing as an alternative to phenotypic testing in a patient with DOCK8 deficiency and severe HSV-1 disease. J Infect Dis. 2020;221:2035-2042. doi:10.1093/infdis/jiaa020
Article PDF
Author and Disclosure Information

Dr. Hopkins is from the Department of Dermatology, Baylor College of Medicine, Houston, Texas. Dr. Lowe is from the Department of Dermatology, Intermountain Medical Group, Salt Lake City, Utah. Dr. Lee is from the Department of Pediatric Dermatology, Texas Children’s Hospital, Houston.

The authors report no conflict of interest.

The eTable is available in the Appendix online at www.mdedge.com/dermatology.

Correspondence: Grace Lee, MD, Texas Children’s Hospital, Department of Pediatric Dermatology, 6701 Fannin St, Mark Wallace Tower, 8th Floor, Houston, TX 77030 ([email protected]).

Issue
Cutis - 108(4)
Publications
Topics
Page Number
218-e4
Sections
Author and Disclosure Information

Dr. Hopkins is from the Department of Dermatology, Baylor College of Medicine, Houston, Texas. Dr. Lowe is from the Department of Dermatology, Intermountain Medical Group, Salt Lake City, Utah. Dr. Lee is from the Department of Pediatric Dermatology, Texas Children’s Hospital, Houston.

The authors report no conflict of interest.

The eTable is available in the Appendix online at www.mdedge.com/dermatology.

Correspondence: Grace Lee, MD, Texas Children’s Hospital, Department of Pediatric Dermatology, 6701 Fannin St, Mark Wallace Tower, 8th Floor, Houston, TX 77030 ([email protected]).

Author and Disclosure Information

Dr. Hopkins is from the Department of Dermatology, Baylor College of Medicine, Houston, Texas. Dr. Lowe is from the Department of Dermatology, Intermountain Medical Group, Salt Lake City, Utah. Dr. Lee is from the Department of Pediatric Dermatology, Texas Children’s Hospital, Houston.

The authors report no conflict of interest.

The eTable is available in the Appendix online at www.mdedge.com/dermatology.

Correspondence: Grace Lee, MD, Texas Children’s Hospital, Department of Pediatric Dermatology, 6701 Fannin St, Mark Wallace Tower, 8th Floor, Houston, TX 77030 ([email protected]).

Article PDF
Article PDF

Dedicator of cytokinesis 8 (DOCK8 ) deficiency is the major cause of autosomal-recessive hyper-IgEsyndrome. 1 Characteristic clinical features including eosinophilia, eczema, and recurrent Staphylococcus aureus cutaneous and respiratory tract infections are common in DOCK8 deficiency, similar to the autosomal-dominant form of hyper-IgE syndrome that is due to defi c iency of signal transducer and activation of transcription 3 (STAT-3 ). 1 In addition, patients with DOCK8 deficiency are particularly susceptible to asthma; food allergies; lymphomas; and severe cutaneous viral infections, including herpes simplex virus (HSV), molluscum contagiosum, varicella-zoster virus, and human papillomavirus. Since the discovery of the DOCK8 gene in 2009, various studies have sought to elucidate the mechanistic contribution of DOCK8 to the dermatologic immune environment. 2 Although cutaneous viral infections such as those caused by HSV typically are short lived and self-limiting in immunocompetent hosts, they have proven to be severe and recalcitrant in the setting of DOCK8 deficiency. 1 Herein, we report the case of a 32-month-old girl with homozygous DOCK8 deficiency who developed acyclovir-resistant cutaneous HSV. 

Case Report

A 32-month-old girl presented with an approximately 2-cm linear erosion along the left posterior auricular sulcus at month 9 of a hospital stay for recurrent infections. Her medical history was notable for multiple upper respiratory tract infections, diffuse eczema, and food allergies. She had presented to an outside hospital at 14 months of age with herpetic gingivostomatitis and eczema herpeticum that was successfully treated with acyclovir. She was readmitted at 20 months of age due to Pneumocystis jiroveci pneumonia, pancytopenia, and disseminated histoplasmosis. Prophylactic oral acyclovir (20 mg/kg twice daily) was started, given her history of HSV infection. Because of recurrent infections, she underwent an immunodeficiency workup. Whole exome sequencing analysis revealed a homozygous deletion c.(528+1_5291)_(1516+1_15171)del in DOCK8 gene–affecting exons 5 to 13. The patient was transferred to our hospital for continued care and as a potential candidate for bone marrow transplant following resolution of the disseminated histoplasmosis infection.

During her hospitalization at the current presentation, she was noted to have a 2-cm linear erosion along the left posterior auricular sulcus. Initial wound care with bacitracin ointment was applied to the area while specimens were obtained and empiric oral acyclovir therapy was initiated (20 mg/kg 4 times daily [QID]), given a clinical impression consistent with cutaneous HSV infection despite acyclovir prophylaxis. Direct immunofluorescence and viral cultures were positive for HSV-1, while bacterial cultures grew methicillin-susceptible S aureus. Cephalexin and mupirocin ointment were started, and acyclovir was continued. After 2 weeks of therapy, there was no visible change in the wound; cultures were repeated, again showing the wound contained HSV. Bacterial cultures this time grew Pseudomonas putida, and the antibiotic regimen was transitioned to cefepime.

After no response to the continued course of therapeutic acyclovir, HSV cultures were sent to the Centers for Disease Control and Prevention for resistance testing, and biopsy of the lesion was performed by the otolaryngology service to rule out malignancy and potential alternative diagnoses. Histopathology showed only reactive inflammation without visible microorganisms on tissue HSV-1/HSV-2 immunostain; however, tissue viral culture was positive for HSV-1. The patient was transitioned back to acyclovir (intravenous [IV] 20 mg/kg QID) with the addition of empiric foscarnet (IV 40 mg/kg 3 times daily) given the worsening appearance of the lesion.  The HSV acyclovir resistance test results from the Centers for Disease Control and Prevention returned soon after and were positive for resistance (median infectious dose, 3.29 µg/L [reference interval, sensitive <2.00 µg/L; resistant >1.90 µg/L]). The patient completed a 21-day course of combination foscarnet and acyclovir therapy, during which time the lesion showed notable improvement and healing. The patient was continued on prophylactic acyclovir (IV 20 mg/kg QID). Unfortunately, the patient eventually died due to complications related to pneumonia.

Comment

Infection in Patients With DOCK8 Deficiency—The gene DOCK8 has emerged as playing a central role in both innate and adaptive immunity, as it is expressed primarily in immune cells and serves as a mediator of numerous processes, including immune synapse formation, cell signaling and trafficking, antibody and cytokine production, and lymphocyte memory.3 Cells that are critical for combating cutaneous viral infections, including skin-resident memory T cells and natural killer cells, are defective, which leads to a severely immunocompromised state in DOCK8-deficient patients with a particular susceptibility to infectious and inflammatory dermatologic disease.4 

Herpes simplex virus infection commonly is seen in DOCK8 deficiency, with retrospective analysis of a DOCK8-deficient cohort revealing HSV infection in approximately 38% of patients.5 Prophylactic acyclovir is essential for DOCK8-deficient individuals with a history of HSV infection given the tendency of the virus to reactivate.6 However, despite prophylaxis, our patient developed an HSV-positive posterior auricular erosion that continued to progress even after increase of the acyclovir dose. Acyclovir resistance testing of the HSV isolated from the wound was positive, confirming the clinical suspicion of the presence of acyclovir-resistant HSV infection.

Acyclovir-Resistant HSV—Acyclovir-resistant HSV in immunosuppressed individuals was first noted in 1982, and most cases since then have occurred in the setting of AIDS and in organ transplant recipients.6 Few reports of acyclovir-resistant HSV in DOCK8 deficiency exist, and to our knowledge, our patient is the youngest DOCK8-deficient individual to be documented with acyclovir-resistant HSV infection.1,7-15 We identified relevant cases from the PubMed and EMBASE databases using the search terms DOCK8 deficiency and acyclovir and DOCK8 deficiency and herpes. The eTable lists other reported cases of acyclovir-resistant HSV in DOCK8-deficient patients. The majority of cases involved school-aged females. Lesion types varied and included herpes labialis, eczema herpeticum, and blepharoconjunctivitis. Escalation of therapy and resolution of the lesion was seen in some cases with administration of subcutaneous pegylated interferon alfa-2b.

 

 

Treatment Alternatives—Acyclovir competitively inhibits viral DNA polymerase by incorporating into elongating viral DNA strands and halting chain synthesis. Acyclovir requires triphosphorylation for activation, and viral thymidine kinase is responsible for the first phosphorylation event. Ninety-five percent of cases of acyclovir resistance are secondary to mutations in viral thymidine kinase. Foscarnet also inhibits viral DNA polymerase but does so directly without the need to be phosphorylated first.6 For this reason, foscarnet often is the drug of choice in the treatment of acyclovir-resistant HSV, as evidenced in our patient. However, foscarnet-resistant HSV strains may develop from mutations in the DNA polymerase gene.

Cidofovir is a nucleotide analogue that requires phosphorylation by host, as opposed to viral, kinases for antiviral activity. Intravenous and topical formulations of cidofovir have proven effective in the treatment of acyclovir- and foscarnet-resistant HSV lesions.6 Cidofovir also can be applied intralesionally, a method that provides targeted therapy and minimizes cidofovir-associated nephrotoxicity.12 Reports of systemic interferon alfa therapy for acyclovir-resistant HSV also exist. A study found IFN-⍺ production by peripheral blood mononuclear cells in DOCK8-deficient individuals to be significantly reduced relative to controls (P<.05).7 There has been complete resolution of acyclovir-resistant HSV lesions with subcutaneous pegylated interferon alfa-2b injections in several DOCK8-deficient patients.7-9

The need for escalating therapy in DOCK8-deficient individuals with acyclovir-resistant HSV infection underscores the essential role of DOCK8 in dermatologic immunity. Our case demonstrates that a high degree of suspicion for cutaneous HSV infection should be adopted in DOCK8-deficient patients of any age, regardless of acyclovir prophylaxis. Viral culture in addition to bacterial cultures should be performed early in patients with cutaneous erosions, and the threshold for HSV resistance testing should be low to minimize morbidity associated with these infections. Early resistance testing in our case could have prevented prolongation of infection and likely eliminated the need for a biopsy.

Conclusion

DOCK8 deficiency presents a unique challenge to dermatologists and other health care providers given the susceptibility of affected individuals to developing a reservoir of severe and potentially resistant viral cutaneous infections. Prophylactic acyclovir may not be sufficient for HSV suppression, even in the youngest of patients, and suspicion for resistance should be high to avoid delays in adequate treatment.

Dedicator of cytokinesis 8 (DOCK8 ) deficiency is the major cause of autosomal-recessive hyper-IgEsyndrome. 1 Characteristic clinical features including eosinophilia, eczema, and recurrent Staphylococcus aureus cutaneous and respiratory tract infections are common in DOCK8 deficiency, similar to the autosomal-dominant form of hyper-IgE syndrome that is due to defi c iency of signal transducer and activation of transcription 3 (STAT-3 ). 1 In addition, patients with DOCK8 deficiency are particularly susceptible to asthma; food allergies; lymphomas; and severe cutaneous viral infections, including herpes simplex virus (HSV), molluscum contagiosum, varicella-zoster virus, and human papillomavirus. Since the discovery of the DOCK8 gene in 2009, various studies have sought to elucidate the mechanistic contribution of DOCK8 to the dermatologic immune environment. 2 Although cutaneous viral infections such as those caused by HSV typically are short lived and self-limiting in immunocompetent hosts, they have proven to be severe and recalcitrant in the setting of DOCK8 deficiency. 1 Herein, we report the case of a 32-month-old girl with homozygous DOCK8 deficiency who developed acyclovir-resistant cutaneous HSV. 

Case Report

A 32-month-old girl presented with an approximately 2-cm linear erosion along the left posterior auricular sulcus at month 9 of a hospital stay for recurrent infections. Her medical history was notable for multiple upper respiratory tract infections, diffuse eczema, and food allergies. She had presented to an outside hospital at 14 months of age with herpetic gingivostomatitis and eczema herpeticum that was successfully treated with acyclovir. She was readmitted at 20 months of age due to Pneumocystis jiroveci pneumonia, pancytopenia, and disseminated histoplasmosis. Prophylactic oral acyclovir (20 mg/kg twice daily) was started, given her history of HSV infection. Because of recurrent infections, she underwent an immunodeficiency workup. Whole exome sequencing analysis revealed a homozygous deletion c.(528+1_5291)_(1516+1_15171)del in DOCK8 gene–affecting exons 5 to 13. The patient was transferred to our hospital for continued care and as a potential candidate for bone marrow transplant following resolution of the disseminated histoplasmosis infection.

During her hospitalization at the current presentation, she was noted to have a 2-cm linear erosion along the left posterior auricular sulcus. Initial wound care with bacitracin ointment was applied to the area while specimens were obtained and empiric oral acyclovir therapy was initiated (20 mg/kg 4 times daily [QID]), given a clinical impression consistent with cutaneous HSV infection despite acyclovir prophylaxis. Direct immunofluorescence and viral cultures were positive for HSV-1, while bacterial cultures grew methicillin-susceptible S aureus. Cephalexin and mupirocin ointment were started, and acyclovir was continued. After 2 weeks of therapy, there was no visible change in the wound; cultures were repeated, again showing the wound contained HSV. Bacterial cultures this time grew Pseudomonas putida, and the antibiotic regimen was transitioned to cefepime.

After no response to the continued course of therapeutic acyclovir, HSV cultures were sent to the Centers for Disease Control and Prevention for resistance testing, and biopsy of the lesion was performed by the otolaryngology service to rule out malignancy and potential alternative diagnoses. Histopathology showed only reactive inflammation without visible microorganisms on tissue HSV-1/HSV-2 immunostain; however, tissue viral culture was positive for HSV-1. The patient was transitioned back to acyclovir (intravenous [IV] 20 mg/kg QID) with the addition of empiric foscarnet (IV 40 mg/kg 3 times daily) given the worsening appearance of the lesion.  The HSV acyclovir resistance test results from the Centers for Disease Control and Prevention returned soon after and were positive for resistance (median infectious dose, 3.29 µg/L [reference interval, sensitive <2.00 µg/L; resistant >1.90 µg/L]). The patient completed a 21-day course of combination foscarnet and acyclovir therapy, during which time the lesion showed notable improvement and healing. The patient was continued on prophylactic acyclovir (IV 20 mg/kg QID). Unfortunately, the patient eventually died due to complications related to pneumonia.

Comment

Infection in Patients With DOCK8 Deficiency—The gene DOCK8 has emerged as playing a central role in both innate and adaptive immunity, as it is expressed primarily in immune cells and serves as a mediator of numerous processes, including immune synapse formation, cell signaling and trafficking, antibody and cytokine production, and lymphocyte memory.3 Cells that are critical for combating cutaneous viral infections, including skin-resident memory T cells and natural killer cells, are defective, which leads to a severely immunocompromised state in DOCK8-deficient patients with a particular susceptibility to infectious and inflammatory dermatologic disease.4 

Herpes simplex virus infection commonly is seen in DOCK8 deficiency, with retrospective analysis of a DOCK8-deficient cohort revealing HSV infection in approximately 38% of patients.5 Prophylactic acyclovir is essential for DOCK8-deficient individuals with a history of HSV infection given the tendency of the virus to reactivate.6 However, despite prophylaxis, our patient developed an HSV-positive posterior auricular erosion that continued to progress even after increase of the acyclovir dose. Acyclovir resistance testing of the HSV isolated from the wound was positive, confirming the clinical suspicion of the presence of acyclovir-resistant HSV infection.

Acyclovir-Resistant HSV—Acyclovir-resistant HSV in immunosuppressed individuals was first noted in 1982, and most cases since then have occurred in the setting of AIDS and in organ transplant recipients.6 Few reports of acyclovir-resistant HSV in DOCK8 deficiency exist, and to our knowledge, our patient is the youngest DOCK8-deficient individual to be documented with acyclovir-resistant HSV infection.1,7-15 We identified relevant cases from the PubMed and EMBASE databases using the search terms DOCK8 deficiency and acyclovir and DOCK8 deficiency and herpes. The eTable lists other reported cases of acyclovir-resistant HSV in DOCK8-deficient patients. The majority of cases involved school-aged females. Lesion types varied and included herpes labialis, eczema herpeticum, and blepharoconjunctivitis. Escalation of therapy and resolution of the lesion was seen in some cases with administration of subcutaneous pegylated interferon alfa-2b.

 

 

Treatment Alternatives—Acyclovir competitively inhibits viral DNA polymerase by incorporating into elongating viral DNA strands and halting chain synthesis. Acyclovir requires triphosphorylation for activation, and viral thymidine kinase is responsible for the first phosphorylation event. Ninety-five percent of cases of acyclovir resistance are secondary to mutations in viral thymidine kinase. Foscarnet also inhibits viral DNA polymerase but does so directly without the need to be phosphorylated first.6 For this reason, foscarnet often is the drug of choice in the treatment of acyclovir-resistant HSV, as evidenced in our patient. However, foscarnet-resistant HSV strains may develop from mutations in the DNA polymerase gene.

Cidofovir is a nucleotide analogue that requires phosphorylation by host, as opposed to viral, kinases for antiviral activity. Intravenous and topical formulations of cidofovir have proven effective in the treatment of acyclovir- and foscarnet-resistant HSV lesions.6 Cidofovir also can be applied intralesionally, a method that provides targeted therapy and minimizes cidofovir-associated nephrotoxicity.12 Reports of systemic interferon alfa therapy for acyclovir-resistant HSV also exist. A study found IFN-⍺ production by peripheral blood mononuclear cells in DOCK8-deficient individuals to be significantly reduced relative to controls (P<.05).7 There has been complete resolution of acyclovir-resistant HSV lesions with subcutaneous pegylated interferon alfa-2b injections in several DOCK8-deficient patients.7-9

The need for escalating therapy in DOCK8-deficient individuals with acyclovir-resistant HSV infection underscores the essential role of DOCK8 in dermatologic immunity. Our case demonstrates that a high degree of suspicion for cutaneous HSV infection should be adopted in DOCK8-deficient patients of any age, regardless of acyclovir prophylaxis. Viral culture in addition to bacterial cultures should be performed early in patients with cutaneous erosions, and the threshold for HSV resistance testing should be low to minimize morbidity associated with these infections. Early resistance testing in our case could have prevented prolongation of infection and likely eliminated the need for a biopsy.

Conclusion

DOCK8 deficiency presents a unique challenge to dermatologists and other health care providers given the susceptibility of affected individuals to developing a reservoir of severe and potentially resistant viral cutaneous infections. Prophylactic acyclovir may not be sufficient for HSV suppression, even in the youngest of patients, and suspicion for resistance should be high to avoid delays in adequate treatment.

References
  1. Chu EY, Freeman AF, Jing H, et al. Cutaneous manifestations of DOCK8 deficiency syndrome. Arch Dermatol. 2012;148:79-84. doi:10.1001/archdermatol.2011.262
  2. Aydin SE, Kilic SS, Aytekin C, et al. DOCK8 deficiency: clinical and immunological phenotype and treatment options—a review of 136 patients. J Clin Immunol. 2015;35:189-198. doi:10.1007/s10875-014-0126-0
  3. Kearney CJ, Randall KL, Oliaro J. DOCK8 regulates signal transduction events to control immunity. Cell Mol Immunol. 2017;14:406-411. doi:10.1038/cmi.2017.9
  4. Zhang Q, Dove CG, Hor JL, et al. DOCK8 regulates lymphocyte shape integrity for skin antiviral immunity. J Exp Med. 2014;211:2549-2566. doi:10.1084/jem.20141307
  5. Engelhardt KR, Gertz EM, Keles S, et al. The extended clinical phenotype of 64 patients with DOCK8 deficiency. J Allergy Clin Immunol. 2015;136:402-412. doi:10.1016/j.jaci.2014.12.1945
  6. Chilukuri S, Rosen T. Management of acyclovir-resistant herpes simplex virus. Dermatol Clin. 2003;21:311-320. doi:10.1016/S0733-8635(02)00093-1
  7. Keles S, Jabara HH, Reisli I, et al. Plasmacytoid dendritic cell depletion in DOCK8 deficiency: rescue of severe herpetic infections with interferon alpha-2b therapy. J Allergy Clin Immunol. 2014;133:1753-1755.e3. doi:10.1016/j.jaci.2014.03.032
  8. Papan C, Hagl B, Heinz V, et al Beneficial IFN-α treatment of tumorous herpes simplex blepharoconjunctivitis in dedicator of cytokinesis 8 deficiency. J Allergy Clin Immunol. 2014;133:1456-1458. doi:10.1016/j.jaci.2014.02.008
  9. Metin A, Kanik-Yuksek S, Ozkaya-Parlakay A, et al. Giant herpes labialis in a child with DOCK8-deficient hyper-IgE syndrome. Pediatr Neonatol. 2016;57:79-80. doi:10.1016/j.pedneo.2015.04.011
  10. Zhang Q, Davis JC, Lamborn IT, et al. Combined immunodeficiency associated with DOCK8 mutations. N Engl J Med. 2009;361:2046-2055. doi:10.1056/NEJMoa0905506
  11. Lei JY, Wang Y, Jaffe ES, et al. Microcystic adnexal carcinoma associated with primary immunodeficiency, recurrent diffuse herpes simplex virus infection, and cutaneous T-cell lymphoma. Am J Dermatopathol. 2000;22:524-529. doi:10.1097/00000372-200012000-00008
  12. Castelo-Soccio L, Bernardin R, Stern J, et al. Successful treatment of acyclovir-resistant herpes simplex virus with intralesional cidofovir. Arch Dermatol. 2010;146:124-126. doi:10.1001/archdermatol.2009.363
  13. Shah NN, Freeman AF, Hickstein DD. Addendum to: haploidentical related donor hematopoietic stem cell transplantation for DOCK8 deficiency using post-transplantation cyclophosphamide. Biol Blood Marrow Transplant. 2019;25:E65-E67. doi:10.1016/j.bbmt.2018.11.014
  14. Freeman AF, Yazigi N, Shah NN, et al. Tandem orthotopic living donor liver transplantation followed by same donor haploidentical hematopoietic stem cell transplantation for DOCK8 deficiency. Transplantation. 2019;103:2144-2149. doi:10.1097/TP.0000000000002649
  15. Casto AM, Stout SC, Selvarangan R, et al. Evaluation of genotypic antiviral resistance testing as an alternative to phenotypic testing in a patient with DOCK8 deficiency and severe HSV-1 disease. J Infect Dis. 2020;221:2035-2042. doi:10.1093/infdis/jiaa020
References
  1. Chu EY, Freeman AF, Jing H, et al. Cutaneous manifestations of DOCK8 deficiency syndrome. Arch Dermatol. 2012;148:79-84. doi:10.1001/archdermatol.2011.262
  2. Aydin SE, Kilic SS, Aytekin C, et al. DOCK8 deficiency: clinical and immunological phenotype and treatment options—a review of 136 patients. J Clin Immunol. 2015;35:189-198. doi:10.1007/s10875-014-0126-0
  3. Kearney CJ, Randall KL, Oliaro J. DOCK8 regulates signal transduction events to control immunity. Cell Mol Immunol. 2017;14:406-411. doi:10.1038/cmi.2017.9
  4. Zhang Q, Dove CG, Hor JL, et al. DOCK8 regulates lymphocyte shape integrity for skin antiviral immunity. J Exp Med. 2014;211:2549-2566. doi:10.1084/jem.20141307
  5. Engelhardt KR, Gertz EM, Keles S, et al. The extended clinical phenotype of 64 patients with DOCK8 deficiency. J Allergy Clin Immunol. 2015;136:402-412. doi:10.1016/j.jaci.2014.12.1945
  6. Chilukuri S, Rosen T. Management of acyclovir-resistant herpes simplex virus. Dermatol Clin. 2003;21:311-320. doi:10.1016/S0733-8635(02)00093-1
  7. Keles S, Jabara HH, Reisli I, et al. Plasmacytoid dendritic cell depletion in DOCK8 deficiency: rescue of severe herpetic infections with interferon alpha-2b therapy. J Allergy Clin Immunol. 2014;133:1753-1755.e3. doi:10.1016/j.jaci.2014.03.032
  8. Papan C, Hagl B, Heinz V, et al Beneficial IFN-α treatment of tumorous herpes simplex blepharoconjunctivitis in dedicator of cytokinesis 8 deficiency. J Allergy Clin Immunol. 2014;133:1456-1458. doi:10.1016/j.jaci.2014.02.008
  9. Metin A, Kanik-Yuksek S, Ozkaya-Parlakay A, et al. Giant herpes labialis in a child with DOCK8-deficient hyper-IgE syndrome. Pediatr Neonatol. 2016;57:79-80. doi:10.1016/j.pedneo.2015.04.011
  10. Zhang Q, Davis JC, Lamborn IT, et al. Combined immunodeficiency associated with DOCK8 mutations. N Engl J Med. 2009;361:2046-2055. doi:10.1056/NEJMoa0905506
  11. Lei JY, Wang Y, Jaffe ES, et al. Microcystic adnexal carcinoma associated with primary immunodeficiency, recurrent diffuse herpes simplex virus infection, and cutaneous T-cell lymphoma. Am J Dermatopathol. 2000;22:524-529. doi:10.1097/00000372-200012000-00008
  12. Castelo-Soccio L, Bernardin R, Stern J, et al. Successful treatment of acyclovir-resistant herpes simplex virus with intralesional cidofovir. Arch Dermatol. 2010;146:124-126. doi:10.1001/archdermatol.2009.363
  13. Shah NN, Freeman AF, Hickstein DD. Addendum to: haploidentical related donor hematopoietic stem cell transplantation for DOCK8 deficiency using post-transplantation cyclophosphamide. Biol Blood Marrow Transplant. 2019;25:E65-E67. doi:10.1016/j.bbmt.2018.11.014
  14. Freeman AF, Yazigi N, Shah NN, et al. Tandem orthotopic living donor liver transplantation followed by same donor haploidentical hematopoietic stem cell transplantation for DOCK8 deficiency. Transplantation. 2019;103:2144-2149. doi:10.1097/TP.0000000000002649
  15. Casto AM, Stout SC, Selvarangan R, et al. Evaluation of genotypic antiviral resistance testing as an alternative to phenotypic testing in a patient with DOCK8 deficiency and severe HSV-1 disease. J Infect Dis. 2020;221:2035-2042. doi:10.1093/infdis/jiaa020
Issue
Cutis - 108(4)
Issue
Cutis - 108(4)
Page Number
218-e4
Page Number
218-e4
Publications
Publications
Topics
Article Type
Display Headline
Acyclovir-Resistant Cutaneous Herpes Simplex Virus in DOCK8 Deficiency  
Display Headline
Acyclovir-Resistant Cutaneous Herpes Simplex Virus in DOCK8 Deficiency  
Sections
Inside the Article

Practice Points

  • Patients with dedicator of cytokinesis 8 ( DOCK 8 ) deficiency are susceptible to development of severe recalcitrant viral cutaneous infections, including herpes simplex virus (HSV).
  • Dermatologists should be aware that prophylactic acyclovir may not be sufficient for HSV suppression in the setting of severe immunodeficiency.
  • Acyclovir-resistant cutaneous HSV lesions require escalation of therapy, which may include addition of foscarnet, cidofovir, or subcutaneous pegylated interferon alfa-2b to the therapeutic regimen.
  • Viral culture should be performed on suspicious lesions in DOCK 8 -deficient patients despite acyclovir prophylaxis, and the threshold for HSV resistance testing should be low.
  •  
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Bullous Amyloidosis Masquerading as Pseudoporphyria

Article Type
Changed
Thu, 10/07/2021 - 07:28
Display Headline
Bullous Amyloidosis Masquerading as Pseudoporphyria

Cutaneous amyloidosis encompasses a variety of clinical presentations. Primary localized cutaneous amyloidosis comprises lichen amyloidosis, macular amyloidosis, and nodular amyloidosis.1 Macular and lichen amyloidosis result from keratin deposits, while nodular amyloidosis results from cutaneous infiltration of plasma cells.2 Primary systemic amyloidosis is due to a plasma cell dyscrasia, particularly multiple myeloma, while secondary systemic amyloidosis occurs in the setting of restrictive cardiomyopathy, congestive heart failure, renal dysfunction, or chronic inflammation, as seen with rheumatoid arthritis, tuberculosis, and various autoinflammatory disorders.2 Plasma cell proliferative disorders are associated with various skin disorders, which may result from aggregated misfolded monoclonal immunoglobulins, indicating light chain–related systemic amyloidosis. Mucocutaneous lesions can occur in 30% to 40% of cases of primary systemic amyloidosis and may present as purpura, ecchymoses, waxy thickening, plaques, subcutaneous nodules, and/or bullae.3,4 When blistering is present, the differential diagnosis is broad and includes autoimmune bullous disease, drug eruptions, enoxaparin-induced bullous hemorrhagic dermatosis, deposition diseases, allergic contact dermatitis, bullous cellulitis, bullous bite reactions, neutrophilic dermatosis, and bullous lichen sclerosus.5 Herein, we present a case of a woman with a bullous skin eruption who eventually was diagnosed with bullous amyloidosis subsequent to a diagnosis of multiple myeloma.

Case Report

A 70-year-old woman presented to our dermatology clinic for evaluation of well-demarcated, hemorrhagic, flaccid vesicles and focal erosions with a rim of erythema on the distal forearms and hands. A shave biopsy from the right forearm showed cell-poor subepidermal vesicular dermatitis. Enzyme-linked immunosorbent assays for bullous pemphigoid antigens 1 and 2 as well as urinary porphyrins were negative. Direct immunofluorescence showed granular IgM at the basement membrane zone around vessels and cytoid bodies. At this time, a preliminary diagnosis of pseudoporphyria was suspected, though no classic medications (eg, nonsteroidal anti-inflammatory drugs, furosemide, antibiotics) or exogenous trigger factors (eg, UV light exposure, dialysis) were temporally related. Three months later, the patient presented with a large hemorrhagic bulla on the distal left forearm (Figure 1) and healing erosions on the dorsal fingers and upper back. Clobetasol ointment was initiated, as an autoimmune bullous dermatosis was suspected.

Large hemorrhagic bulla on the distal left forearm later confirmed to be bullous amyloidosis.
FIGURE 1. Large hemorrhagic bulla on the distal left forearm later confirmed to be bullous amyloidosis.

Approximately 1 year after she was first seen in our outpatient clinic, the patient was hospitalized for induction of chemotherapy—cyclophosphamide, bortezomib, and dexamethasone—for a new diagnosis of stage III multiple myeloma. A workup for back pain revealed multiple compression fractures and a plasma cell neoplasm with elevated λ light chains, which was confirmed with a bone marrow biopsy. During an inpatient dermatology consultation, we noted the development of intraoral hemorrhagic vesicles and worsening generalization of the hemorrhagic bullae, with healing erosions and intact hemorrhagic bullae on the dorsal hands, fingers (Figure 2), and upper back.

A repeat biopsy displayed bullous amyloidosis. Histopathologic examination revealed an ulcerated subepidermal blister with fibrin deposition at the ulcer base. A periadnexal, scant, eosinophilic deposition with extravasated red blood cells was appreciated. Amorphous eosinophilic deposits were found within the detached fragment of the epidermis and inflammatory infiltrate. A Congo red stain highlighted these areas with a salmon pink–colored material. Congo red staining showed a moderate amount of pale, apple green, birefringent deposit within these areas on polarized light examination.

A few months later, the patient was re-admitted, and the amount of skin detachment prompted the primary team to ask for another consultation. Although the extensive skin sloughing resembled toxic epidermal necrolysis, a repeat biopsy confirmed bullous amyloidosis.

Comment

Amyloidosis Histopathology—Amyloidoses represent a wide array of disorders with deposition of β-pleated sheets or amyloid fibrils, often with cutaneous manifestations.2,3 Primary systemic amyloidosis has been associated with underlying dyscrasia or multiple myeloma.6 In such cases, the skin lesions of multiple myeloma may result from a collection of misfolded monoclonal immunoglobulins or their fragments, as in light chain–related systemic amyloidosis.3 Histopathologically, both systemic and cutaneous amyloidosis appear similar and display deposition of amorphous, eosinophilic, fissured amyloid material in the dermis. Congo red stains the material orange-red and will display a characteristic apple green birefringence under polarized light.4 Although bullous amyloid lesions are rare, the cutaneous forms of these lesions can be an important sign of plasma cell dyscrasia.7

Presentation of Bullous Amyloidosis—Bullous manifestations rarely have been noted in the primary cutaneous forms of amyloidosis.5,8,9 Importantly, cutaneous blistering more often is linked to systemic forms of amyloidosis with multiorgan involvement, including primary systemic and myeloma-associated amyloidosis.5,10 However, patients with localized bullous cutaneous amyloidosis without systemic involvement also have been seen.10,11 Bullae may occur at any time, with contents that frequently are hemorrhagic due to capillary fragility.12,13 Bullous manifestations raise the differential diagnoses of bullous pemphigoid, epidermolysis bullosa acquisita, linear IgA disease, porphyria cutanea tarda, pseudoporphyria, bullous drug eruption, bullous eruption of renal dialysis, or bullous lupus erythematosus.5,13-17

In our patient, the acral distribution of bullae, presence of hemorrhage, chronicity of symptoms, and negative enzyme-linked immunosorbent assay initially suggested a diagnosis of pseudoporphyria. However, the presence of intraoral hemorrhagic vesicles and subsequent confirmatory pathology aided in differentiating bullous amyloidosis from pseudoporphyria. Nodular localized primary cutaneous amyloidosis, a rare form of skin-restricted amyloidoses, can coexist with bullous lesions. Of note, reported cases of nodular localized primary cutaneous amyloidosis did not result in development of multiple myeloma.5,10

Bullae are located either subepidermally or intradermally, and bullous lesions of cutaneous amyloidosis typically demonstrate subepidermal or superficial intradermal clefting on light microscopy.5,10,12 Histopathology of bullous amyloidosis shows intradermal or subepidermal blister formation and amorphous eosinophilic material showing apple green birefringence with Congo red staining deposited in the dermis and/or around the adipocytes and blood vessel walls.12,18-20 In prior cases, direct immunofluorescence of bullous amyloidosis revealed absent immunoglobulin (IgG, IgA, IgM) or complement (C3 and C9) deposits in the basement membrane zone or dermis.13,21,22 In these cases, electron microscopy was useful in diagnosis, as it showed the presence of amyloid deposits.21,22

Cause of Bullae—Various mechanisms are thought to trigger the blister formation in amyloidosis. Bullae created from trauma or friction often present as tense painful blisters that commonly are hemorrhagic.10,23 Amyloid deposits in the walls of blood vessels and the affinity of dermal amyloid in blood vessel walls to surrounding collagen likely leads to increased fragility of capillaries and the dermal matrix, hemorrhagic tendency, and infrapapillary blisters, thus creating hemorrhagic bullous eruptions.24,25 Specifically, close proximity of immunoglobulin-derived amyloid oligomers to epidermal keratinocytes may be toxic and therefore could trigger subepidermal bullous change.5 Additionally, alteration in the physicochemical properties of the amyloidal protein might explain bullous eruption.9 Trauma or rubbing of the hands and feet may precipitate the acral blister formation in bullous amyloidosis.5,11

Due to deposition of these amyloid fibrils, skin bleeding in these patients is called amyloid or pinch purpura. Vessel wall fragility and damage by amyloid are the principal causes of periorbital and gastrointestinal tract bleeding.26 Destruction of the lamina densa and widening of the intercellular space between keratinocytes by amyloid globules induce skin fragility.11

Although uncommon, various cases of bullous amyloidosis have been reported in the literature. Multiple myeloma patients represent the majority of those reported to have bullous amyloidosis.6,7,13,24,27-30 Plasmacytoma-associated bullous amyloid purpura and paraproteinemia also have been noted.25 Multiple myeloma with secondary AL amyloidosis has been seen with amyloid purpura and atraumatic ecchymoses of the face, highlighting the hemorrhage noted in these patients.26

Management of Amyloidosis—Various treatment options have been attempted for primary cutaneous amyloidosis, including oral retinoids, corticosteroids, cyclophosphamide, cyclosporine, amitriptyline, colchicine, cepharanthin, tacrolimus, dimethyl sulfoxide, vitamin D3 analogs, capsaicin, menthol, hydrocolloid dressings, surgical modalities, laser treatment, and phototherapy.1 There is no clear consensus for therapeutic modalities except for treating the underlying plasma cell dyscrasia in primary systemic amyloidosis.

Conclusion

We report the case of a patient displaying signs of pseudoporphyria that ultimately proved to be bullous amyloidosis, or what we termed pseudopseudoporphyria. Bullous amyloidosis should be considered in the differential diagnoses of hemorrhagic bullous skin eruptions. Particular attention should be given to a systemic workup for multiple myeloma when hemorrhagic vesicles/bullae are chronic and coexist with purpura, angina bullosa hemorrhagica, fatigue/weight loss, and/or macroglossia.

References
  1. Weidner T, Illing T, Elsner P. Primary localized cutaneous amyloidosis: a systematic treatment review. Am J Clin Dermatol. 2017;18:629-642.
  2. Bolognia JL, Schaffer JV, Duncan KO, et al. Amyloidosis. Dermatology Essentials. Elsevier Saunders; 2014:341-345.
  3. Bhutani M, Shahid Z, Schnebelen A, et al. Cutaneous manifestations of multiple myeloma and other plasma cell proliferative disorders. Semin Oncol. 2016;43:395-400.
  4. Terushkin V, Boyd KP, Patel RR, et al. Primary localized cutaneous amyloidosis. Dermatol Online J. 2013;19:20711.
  5. LaChance A, Phelps A, Finch J, et al. Nodular localized primary cutaneous amyloidosis: a bullous variant. Clin Exp Dermatol. 2014;39:344-347.
  6. Gonzalez-Ramos J, Garrido-Gutiérrez C, González-Silva Y, et al. Relapsing bullous amyloidosis of the oral mucosa and acquired cutis laxa in a patient with multiple myeloma: a rare triple association. Clin Exp Dermatol. 2017;42:410-412.
  7. Kanoh T. Bullous amyloidosis [in Japanese]. Rinsho Ketsueki. 1993;34:1050-1052.
  8. Johnson TM, Rapini RP, Hebert AA, et al. Bullous amyloidosis. Cutis. 1989;43:346-352.
  9. Houman MH, Smiti KM, Ben Ghorbel I, et al. Bullous amyloidosis. Ann Dermatol Venereol. 2002;129:299-302.
  10. Sanusi T, Li Y, Qian Y, et al. Primary localized cutaneous nodular amyloidosis with bullous lesions. Indian J Dermatol Venereol Leprol. 2015;81:400-402.
  11. Ochiai T, Morishima T, Hao T, et al. Bullous amyloidosis: the mechanism of blister formation revealed by electron microscopy. J Cutan Pathol. 2001;28:407-411.
  12. Chu CH, Chan JY, Hsieh SW, et al. Diffuse ecchymoses and blisters on a yellowish waxy base: a case of bullous amyloidosis. J Dermatol. 2016;43:713-714.
  13. Wang XD, Shen H, Liu ZH. Diffuse haemorrhagic bullous amyloidosis with multiple myeloma. Clin Exp Dermatol. 2008;33:94-96.
  14. Biswas P, Aggarwal I, Sen D, et al. Bullous pemphigoid clinically presenting as lichen amyloidosis. Indian J Dermatol Venereol Leprol. 2014;80:544-546.
  15. Bluhm JF 3rd. Bullous dermatosis vs amyloidosis. Arch Dermatol. 1981;117:252.
  16. Bluhm JF 3rd. Bullous amyloidosis vs epidermolysis bullosa acquisita. JAMA. 1981;245:32.
  17. Murphy GM, Wright J, Nicholls DS, et al. Sunbed-induced pseudoporphyria. Br J Dermatol. 1989;120:555-562.
  18. Pramatarov K, Lazarova A, Mateev G, et al. Bullous hemorrhagic primary systemic amyloidosis. Int J Dermatol. 1990;29:211-213.
  19. Bieber T, Ruzicka T, Linke RP, et al. Hemorrhagic bullous amyloidosis. a histologic, immunocytochemical, and ultrastructural study of two patients. Arch Dermatol. 1988;124:1683-1686.
  20. Khoo BP, Tay YK. Lichen amyloidosis: a bullous variant. Ann Acad Med Singapore. 2000;29:105-107.
  21. Asahina A, Hasegawa K, Ishiyama M, et al. Bullous amyloidosis mimicking bullous pemphigoid: usefulness of electron microscopic examination. Acta Derm Venereol. 2010;90:427-428.
  22. Schmutz JL, Barbaud A, Cuny JF, et al. Bullous amyloidosis [in French]. Ann Dermatol Venereol. 1988;115:295-301.
  23. Lachmann HJ, Hawkins PN. Amyloidosis of the skin. In: Goldsmith LA, Katz SI, Gilchrest BA, et al, eds. Fitzpatrick’s Dermatology in General Medicine. 8th ed. McGraw-Hill; 2012:1574-1583.
  24. Grundmann JU, Bonnekoh B, Gollnick H. Extensive haemorrhagic-bullous skin manifestation of systemic AA-amyloidosis associated with IgG lambda-myeloma. Eur J Dermatol. 2000;10:139-142.
  25. Hödl S, Turek TD, Kerl H. Plasmocytoma-associated bullous hemorrhagic amyloidosis of the skin [in German]. Hautarzt. 1982;33:556-558.
  26. Colucci G, Alberio L, Demarmels Biasiutti F, et al. Bilateral periorbital ecchymoses. an often missed sign of amyloid purpura. Hamostaseologie. 2014;34:249-252.
  27. Behera B, Pattnaik M, Sahu B, et al. Cutaneous manifestations of multiple myeloma. Indian J Dermatol. 2016;61:668-671.
  28. Fujita Y, Tsuji-Abe Y, Sato-Matsumura KC, et al. Nail dystrophy and blisters as sole manifestations in myeloma-associated amyloidosis. J Am Acad Dermatol. 2006;54:712-714.
  29. Chang SL, Lai PC, Cheng CJ, et al. Bullous amyloidosis in a hemodialysis patient is myeloma-associated rather than hemodialysis-associated amyloidosis. Amyloid. 2007;14:153-156.
  30. Winzer M, Ruppert M, Baretton G, et al. Bullous poikilodermatitic amyloidosis of the skin with junctional bulla development in IgG light chain plasmacytoma of the lambda type. histology, immunohistology and electron microscopy [in German]. Hautarzt. 1992;43:199-204.
Article PDF
Author and Disclosure Information

Dr. Darji is from the Department of Dermatology, Saint Louis University, Missouri. Dr. Butala is from the Dermatology Department, Lancaster Medical Offices, Kaiser Permanente, California. Drs. Manders and Green are from the Department of Dermatology, Cooper University Health Care, Camden, New Jersey.

The authors report no conflict of interest.

Correspondence: Justin J. Green, MD, Cooper University Health Care, Department of Dermatology, 3 Cooper Plaza, Ste 211, Camden, NJ 08103 ([email protected]).

Issue
cutis - 108(3)
Publications
Topics
Page Number
e25-e28
Sections
Author and Disclosure Information

Dr. Darji is from the Department of Dermatology, Saint Louis University, Missouri. Dr. Butala is from the Dermatology Department, Lancaster Medical Offices, Kaiser Permanente, California. Drs. Manders and Green are from the Department of Dermatology, Cooper University Health Care, Camden, New Jersey.

The authors report no conflict of interest.

Correspondence: Justin J. Green, MD, Cooper University Health Care, Department of Dermatology, 3 Cooper Plaza, Ste 211, Camden, NJ 08103 ([email protected]).

Author and Disclosure Information

Dr. Darji is from the Department of Dermatology, Saint Louis University, Missouri. Dr. Butala is from the Dermatology Department, Lancaster Medical Offices, Kaiser Permanente, California. Drs. Manders and Green are from the Department of Dermatology, Cooper University Health Care, Camden, New Jersey.

The authors report no conflict of interest.

Correspondence: Justin J. Green, MD, Cooper University Health Care, Department of Dermatology, 3 Cooper Plaza, Ste 211, Camden, NJ 08103 ([email protected]).

Article PDF
Article PDF

Cutaneous amyloidosis encompasses a variety of clinical presentations. Primary localized cutaneous amyloidosis comprises lichen amyloidosis, macular amyloidosis, and nodular amyloidosis.1 Macular and lichen amyloidosis result from keratin deposits, while nodular amyloidosis results from cutaneous infiltration of plasma cells.2 Primary systemic amyloidosis is due to a plasma cell dyscrasia, particularly multiple myeloma, while secondary systemic amyloidosis occurs in the setting of restrictive cardiomyopathy, congestive heart failure, renal dysfunction, or chronic inflammation, as seen with rheumatoid arthritis, tuberculosis, and various autoinflammatory disorders.2 Plasma cell proliferative disorders are associated with various skin disorders, which may result from aggregated misfolded monoclonal immunoglobulins, indicating light chain–related systemic amyloidosis. Mucocutaneous lesions can occur in 30% to 40% of cases of primary systemic amyloidosis and may present as purpura, ecchymoses, waxy thickening, plaques, subcutaneous nodules, and/or bullae.3,4 When blistering is present, the differential diagnosis is broad and includes autoimmune bullous disease, drug eruptions, enoxaparin-induced bullous hemorrhagic dermatosis, deposition diseases, allergic contact dermatitis, bullous cellulitis, bullous bite reactions, neutrophilic dermatosis, and bullous lichen sclerosus.5 Herein, we present a case of a woman with a bullous skin eruption who eventually was diagnosed with bullous amyloidosis subsequent to a diagnosis of multiple myeloma.

Case Report

A 70-year-old woman presented to our dermatology clinic for evaluation of well-demarcated, hemorrhagic, flaccid vesicles and focal erosions with a rim of erythema on the distal forearms and hands. A shave biopsy from the right forearm showed cell-poor subepidermal vesicular dermatitis. Enzyme-linked immunosorbent assays for bullous pemphigoid antigens 1 and 2 as well as urinary porphyrins were negative. Direct immunofluorescence showed granular IgM at the basement membrane zone around vessels and cytoid bodies. At this time, a preliminary diagnosis of pseudoporphyria was suspected, though no classic medications (eg, nonsteroidal anti-inflammatory drugs, furosemide, antibiotics) or exogenous trigger factors (eg, UV light exposure, dialysis) were temporally related. Three months later, the patient presented with a large hemorrhagic bulla on the distal left forearm (Figure 1) and healing erosions on the dorsal fingers and upper back. Clobetasol ointment was initiated, as an autoimmune bullous dermatosis was suspected.

Large hemorrhagic bulla on the distal left forearm later confirmed to be bullous amyloidosis.
FIGURE 1. Large hemorrhagic bulla on the distal left forearm later confirmed to be bullous amyloidosis.

Approximately 1 year after she was first seen in our outpatient clinic, the patient was hospitalized for induction of chemotherapy—cyclophosphamide, bortezomib, and dexamethasone—for a new diagnosis of stage III multiple myeloma. A workup for back pain revealed multiple compression fractures and a plasma cell neoplasm with elevated λ light chains, which was confirmed with a bone marrow biopsy. During an inpatient dermatology consultation, we noted the development of intraoral hemorrhagic vesicles and worsening generalization of the hemorrhagic bullae, with healing erosions and intact hemorrhagic bullae on the dorsal hands, fingers (Figure 2), and upper back.

A repeat biopsy displayed bullous amyloidosis. Histopathologic examination revealed an ulcerated subepidermal blister with fibrin deposition at the ulcer base. A periadnexal, scant, eosinophilic deposition with extravasated red blood cells was appreciated. Amorphous eosinophilic deposits were found within the detached fragment of the epidermis and inflammatory infiltrate. A Congo red stain highlighted these areas with a salmon pink–colored material. Congo red staining showed a moderate amount of pale, apple green, birefringent deposit within these areas on polarized light examination.

A few months later, the patient was re-admitted, and the amount of skin detachment prompted the primary team to ask for another consultation. Although the extensive skin sloughing resembled toxic epidermal necrolysis, a repeat biopsy confirmed bullous amyloidosis.

Comment

Amyloidosis Histopathology—Amyloidoses represent a wide array of disorders with deposition of β-pleated sheets or amyloid fibrils, often with cutaneous manifestations.2,3 Primary systemic amyloidosis has been associated with underlying dyscrasia or multiple myeloma.6 In such cases, the skin lesions of multiple myeloma may result from a collection of misfolded monoclonal immunoglobulins or their fragments, as in light chain–related systemic amyloidosis.3 Histopathologically, both systemic and cutaneous amyloidosis appear similar and display deposition of amorphous, eosinophilic, fissured amyloid material in the dermis. Congo red stains the material orange-red and will display a characteristic apple green birefringence under polarized light.4 Although bullous amyloid lesions are rare, the cutaneous forms of these lesions can be an important sign of plasma cell dyscrasia.7

Presentation of Bullous Amyloidosis—Bullous manifestations rarely have been noted in the primary cutaneous forms of amyloidosis.5,8,9 Importantly, cutaneous blistering more often is linked to systemic forms of amyloidosis with multiorgan involvement, including primary systemic and myeloma-associated amyloidosis.5,10 However, patients with localized bullous cutaneous amyloidosis without systemic involvement also have been seen.10,11 Bullae may occur at any time, with contents that frequently are hemorrhagic due to capillary fragility.12,13 Bullous manifestations raise the differential diagnoses of bullous pemphigoid, epidermolysis bullosa acquisita, linear IgA disease, porphyria cutanea tarda, pseudoporphyria, bullous drug eruption, bullous eruption of renal dialysis, or bullous lupus erythematosus.5,13-17

In our patient, the acral distribution of bullae, presence of hemorrhage, chronicity of symptoms, and negative enzyme-linked immunosorbent assay initially suggested a diagnosis of pseudoporphyria. However, the presence of intraoral hemorrhagic vesicles and subsequent confirmatory pathology aided in differentiating bullous amyloidosis from pseudoporphyria. Nodular localized primary cutaneous amyloidosis, a rare form of skin-restricted amyloidoses, can coexist with bullous lesions. Of note, reported cases of nodular localized primary cutaneous amyloidosis did not result in development of multiple myeloma.5,10

Bullae are located either subepidermally or intradermally, and bullous lesions of cutaneous amyloidosis typically demonstrate subepidermal or superficial intradermal clefting on light microscopy.5,10,12 Histopathology of bullous amyloidosis shows intradermal or subepidermal blister formation and amorphous eosinophilic material showing apple green birefringence with Congo red staining deposited in the dermis and/or around the adipocytes and blood vessel walls.12,18-20 In prior cases, direct immunofluorescence of bullous amyloidosis revealed absent immunoglobulin (IgG, IgA, IgM) or complement (C3 and C9) deposits in the basement membrane zone or dermis.13,21,22 In these cases, electron microscopy was useful in diagnosis, as it showed the presence of amyloid deposits.21,22

Cause of Bullae—Various mechanisms are thought to trigger the blister formation in amyloidosis. Bullae created from trauma or friction often present as tense painful blisters that commonly are hemorrhagic.10,23 Amyloid deposits in the walls of blood vessels and the affinity of dermal amyloid in blood vessel walls to surrounding collagen likely leads to increased fragility of capillaries and the dermal matrix, hemorrhagic tendency, and infrapapillary blisters, thus creating hemorrhagic bullous eruptions.24,25 Specifically, close proximity of immunoglobulin-derived amyloid oligomers to epidermal keratinocytes may be toxic and therefore could trigger subepidermal bullous change.5 Additionally, alteration in the physicochemical properties of the amyloidal protein might explain bullous eruption.9 Trauma or rubbing of the hands and feet may precipitate the acral blister formation in bullous amyloidosis.5,11

Due to deposition of these amyloid fibrils, skin bleeding in these patients is called amyloid or pinch purpura. Vessel wall fragility and damage by amyloid are the principal causes of periorbital and gastrointestinal tract bleeding.26 Destruction of the lamina densa and widening of the intercellular space between keratinocytes by amyloid globules induce skin fragility.11

Although uncommon, various cases of bullous amyloidosis have been reported in the literature. Multiple myeloma patients represent the majority of those reported to have bullous amyloidosis.6,7,13,24,27-30 Plasmacytoma-associated bullous amyloid purpura and paraproteinemia also have been noted.25 Multiple myeloma with secondary AL amyloidosis has been seen with amyloid purpura and atraumatic ecchymoses of the face, highlighting the hemorrhage noted in these patients.26

Management of Amyloidosis—Various treatment options have been attempted for primary cutaneous amyloidosis, including oral retinoids, corticosteroids, cyclophosphamide, cyclosporine, amitriptyline, colchicine, cepharanthin, tacrolimus, dimethyl sulfoxide, vitamin D3 analogs, capsaicin, menthol, hydrocolloid dressings, surgical modalities, laser treatment, and phototherapy.1 There is no clear consensus for therapeutic modalities except for treating the underlying plasma cell dyscrasia in primary systemic amyloidosis.

Conclusion

We report the case of a patient displaying signs of pseudoporphyria that ultimately proved to be bullous amyloidosis, or what we termed pseudopseudoporphyria. Bullous amyloidosis should be considered in the differential diagnoses of hemorrhagic bullous skin eruptions. Particular attention should be given to a systemic workup for multiple myeloma when hemorrhagic vesicles/bullae are chronic and coexist with purpura, angina bullosa hemorrhagica, fatigue/weight loss, and/or macroglossia.

Cutaneous amyloidosis encompasses a variety of clinical presentations. Primary localized cutaneous amyloidosis comprises lichen amyloidosis, macular amyloidosis, and nodular amyloidosis.1 Macular and lichen amyloidosis result from keratin deposits, while nodular amyloidosis results from cutaneous infiltration of plasma cells.2 Primary systemic amyloidosis is due to a plasma cell dyscrasia, particularly multiple myeloma, while secondary systemic amyloidosis occurs in the setting of restrictive cardiomyopathy, congestive heart failure, renal dysfunction, or chronic inflammation, as seen with rheumatoid arthritis, tuberculosis, and various autoinflammatory disorders.2 Plasma cell proliferative disorders are associated with various skin disorders, which may result from aggregated misfolded monoclonal immunoglobulins, indicating light chain–related systemic amyloidosis. Mucocutaneous lesions can occur in 30% to 40% of cases of primary systemic amyloidosis and may present as purpura, ecchymoses, waxy thickening, plaques, subcutaneous nodules, and/or bullae.3,4 When blistering is present, the differential diagnosis is broad and includes autoimmune bullous disease, drug eruptions, enoxaparin-induced bullous hemorrhagic dermatosis, deposition diseases, allergic contact dermatitis, bullous cellulitis, bullous bite reactions, neutrophilic dermatosis, and bullous lichen sclerosus.5 Herein, we present a case of a woman with a bullous skin eruption who eventually was diagnosed with bullous amyloidosis subsequent to a diagnosis of multiple myeloma.

Case Report

A 70-year-old woman presented to our dermatology clinic for evaluation of well-demarcated, hemorrhagic, flaccid vesicles and focal erosions with a rim of erythema on the distal forearms and hands. A shave biopsy from the right forearm showed cell-poor subepidermal vesicular dermatitis. Enzyme-linked immunosorbent assays for bullous pemphigoid antigens 1 and 2 as well as urinary porphyrins were negative. Direct immunofluorescence showed granular IgM at the basement membrane zone around vessels and cytoid bodies. At this time, a preliminary diagnosis of pseudoporphyria was suspected, though no classic medications (eg, nonsteroidal anti-inflammatory drugs, furosemide, antibiotics) or exogenous trigger factors (eg, UV light exposure, dialysis) were temporally related. Three months later, the patient presented with a large hemorrhagic bulla on the distal left forearm (Figure 1) and healing erosions on the dorsal fingers and upper back. Clobetasol ointment was initiated, as an autoimmune bullous dermatosis was suspected.

Large hemorrhagic bulla on the distal left forearm later confirmed to be bullous amyloidosis.
FIGURE 1. Large hemorrhagic bulla on the distal left forearm later confirmed to be bullous amyloidosis.

Approximately 1 year after she was first seen in our outpatient clinic, the patient was hospitalized for induction of chemotherapy—cyclophosphamide, bortezomib, and dexamethasone—for a new diagnosis of stage III multiple myeloma. A workup for back pain revealed multiple compression fractures and a plasma cell neoplasm with elevated λ light chains, which was confirmed with a bone marrow biopsy. During an inpatient dermatology consultation, we noted the development of intraoral hemorrhagic vesicles and worsening generalization of the hemorrhagic bullae, with healing erosions and intact hemorrhagic bullae on the dorsal hands, fingers (Figure 2), and upper back.

A repeat biopsy displayed bullous amyloidosis. Histopathologic examination revealed an ulcerated subepidermal blister with fibrin deposition at the ulcer base. A periadnexal, scant, eosinophilic deposition with extravasated red blood cells was appreciated. Amorphous eosinophilic deposits were found within the detached fragment of the epidermis and inflammatory infiltrate. A Congo red stain highlighted these areas with a salmon pink–colored material. Congo red staining showed a moderate amount of pale, apple green, birefringent deposit within these areas on polarized light examination.

A few months later, the patient was re-admitted, and the amount of skin detachment prompted the primary team to ask for another consultation. Although the extensive skin sloughing resembled toxic epidermal necrolysis, a repeat biopsy confirmed bullous amyloidosis.

Comment

Amyloidosis Histopathology—Amyloidoses represent a wide array of disorders with deposition of β-pleated sheets or amyloid fibrils, often with cutaneous manifestations.2,3 Primary systemic amyloidosis has been associated with underlying dyscrasia or multiple myeloma.6 In such cases, the skin lesions of multiple myeloma may result from a collection of misfolded monoclonal immunoglobulins or their fragments, as in light chain–related systemic amyloidosis.3 Histopathologically, both systemic and cutaneous amyloidosis appear similar and display deposition of amorphous, eosinophilic, fissured amyloid material in the dermis. Congo red stains the material orange-red and will display a characteristic apple green birefringence under polarized light.4 Although bullous amyloid lesions are rare, the cutaneous forms of these lesions can be an important sign of plasma cell dyscrasia.7

Presentation of Bullous Amyloidosis—Bullous manifestations rarely have been noted in the primary cutaneous forms of amyloidosis.5,8,9 Importantly, cutaneous blistering more often is linked to systemic forms of amyloidosis with multiorgan involvement, including primary systemic and myeloma-associated amyloidosis.5,10 However, patients with localized bullous cutaneous amyloidosis without systemic involvement also have been seen.10,11 Bullae may occur at any time, with contents that frequently are hemorrhagic due to capillary fragility.12,13 Bullous manifestations raise the differential diagnoses of bullous pemphigoid, epidermolysis bullosa acquisita, linear IgA disease, porphyria cutanea tarda, pseudoporphyria, bullous drug eruption, bullous eruption of renal dialysis, or bullous lupus erythematosus.5,13-17

In our patient, the acral distribution of bullae, presence of hemorrhage, chronicity of symptoms, and negative enzyme-linked immunosorbent assay initially suggested a diagnosis of pseudoporphyria. However, the presence of intraoral hemorrhagic vesicles and subsequent confirmatory pathology aided in differentiating bullous amyloidosis from pseudoporphyria. Nodular localized primary cutaneous amyloidosis, a rare form of skin-restricted amyloidoses, can coexist with bullous lesions. Of note, reported cases of nodular localized primary cutaneous amyloidosis did not result in development of multiple myeloma.5,10

Bullae are located either subepidermally or intradermally, and bullous lesions of cutaneous amyloidosis typically demonstrate subepidermal or superficial intradermal clefting on light microscopy.5,10,12 Histopathology of bullous amyloidosis shows intradermal or subepidermal blister formation and amorphous eosinophilic material showing apple green birefringence with Congo red staining deposited in the dermis and/or around the adipocytes and blood vessel walls.12,18-20 In prior cases, direct immunofluorescence of bullous amyloidosis revealed absent immunoglobulin (IgG, IgA, IgM) or complement (C3 and C9) deposits in the basement membrane zone or dermis.13,21,22 In these cases, electron microscopy was useful in diagnosis, as it showed the presence of amyloid deposits.21,22

Cause of Bullae—Various mechanisms are thought to trigger the blister formation in amyloidosis. Bullae created from trauma or friction often present as tense painful blisters that commonly are hemorrhagic.10,23 Amyloid deposits in the walls of blood vessels and the affinity of dermal amyloid in blood vessel walls to surrounding collagen likely leads to increased fragility of capillaries and the dermal matrix, hemorrhagic tendency, and infrapapillary blisters, thus creating hemorrhagic bullous eruptions.24,25 Specifically, close proximity of immunoglobulin-derived amyloid oligomers to epidermal keratinocytes may be toxic and therefore could trigger subepidermal bullous change.5 Additionally, alteration in the physicochemical properties of the amyloidal protein might explain bullous eruption.9 Trauma or rubbing of the hands and feet may precipitate the acral blister formation in bullous amyloidosis.5,11

Due to deposition of these amyloid fibrils, skin bleeding in these patients is called amyloid or pinch purpura. Vessel wall fragility and damage by amyloid are the principal causes of periorbital and gastrointestinal tract bleeding.26 Destruction of the lamina densa and widening of the intercellular space between keratinocytes by amyloid globules induce skin fragility.11

Although uncommon, various cases of bullous amyloidosis have been reported in the literature. Multiple myeloma patients represent the majority of those reported to have bullous amyloidosis.6,7,13,24,27-30 Plasmacytoma-associated bullous amyloid purpura and paraproteinemia also have been noted.25 Multiple myeloma with secondary AL amyloidosis has been seen with amyloid purpura and atraumatic ecchymoses of the face, highlighting the hemorrhage noted in these patients.26

Management of Amyloidosis—Various treatment options have been attempted for primary cutaneous amyloidosis, including oral retinoids, corticosteroids, cyclophosphamide, cyclosporine, amitriptyline, colchicine, cepharanthin, tacrolimus, dimethyl sulfoxide, vitamin D3 analogs, capsaicin, menthol, hydrocolloid dressings, surgical modalities, laser treatment, and phototherapy.1 There is no clear consensus for therapeutic modalities except for treating the underlying plasma cell dyscrasia in primary systemic amyloidosis.

Conclusion

We report the case of a patient displaying signs of pseudoporphyria that ultimately proved to be bullous amyloidosis, or what we termed pseudopseudoporphyria. Bullous amyloidosis should be considered in the differential diagnoses of hemorrhagic bullous skin eruptions. Particular attention should be given to a systemic workup for multiple myeloma when hemorrhagic vesicles/bullae are chronic and coexist with purpura, angina bullosa hemorrhagica, fatigue/weight loss, and/or macroglossia.

References
  1. Weidner T, Illing T, Elsner P. Primary localized cutaneous amyloidosis: a systematic treatment review. Am J Clin Dermatol. 2017;18:629-642.
  2. Bolognia JL, Schaffer JV, Duncan KO, et al. Amyloidosis. Dermatology Essentials. Elsevier Saunders; 2014:341-345.
  3. Bhutani M, Shahid Z, Schnebelen A, et al. Cutaneous manifestations of multiple myeloma and other plasma cell proliferative disorders. Semin Oncol. 2016;43:395-400.
  4. Terushkin V, Boyd KP, Patel RR, et al. Primary localized cutaneous amyloidosis. Dermatol Online J. 2013;19:20711.
  5. LaChance A, Phelps A, Finch J, et al. Nodular localized primary cutaneous amyloidosis: a bullous variant. Clin Exp Dermatol. 2014;39:344-347.
  6. Gonzalez-Ramos J, Garrido-Gutiérrez C, González-Silva Y, et al. Relapsing bullous amyloidosis of the oral mucosa and acquired cutis laxa in a patient with multiple myeloma: a rare triple association. Clin Exp Dermatol. 2017;42:410-412.
  7. Kanoh T. Bullous amyloidosis [in Japanese]. Rinsho Ketsueki. 1993;34:1050-1052.
  8. Johnson TM, Rapini RP, Hebert AA, et al. Bullous amyloidosis. Cutis. 1989;43:346-352.
  9. Houman MH, Smiti KM, Ben Ghorbel I, et al. Bullous amyloidosis. Ann Dermatol Venereol. 2002;129:299-302.
  10. Sanusi T, Li Y, Qian Y, et al. Primary localized cutaneous nodular amyloidosis with bullous lesions. Indian J Dermatol Venereol Leprol. 2015;81:400-402.
  11. Ochiai T, Morishima T, Hao T, et al. Bullous amyloidosis: the mechanism of blister formation revealed by electron microscopy. J Cutan Pathol. 2001;28:407-411.
  12. Chu CH, Chan JY, Hsieh SW, et al. Diffuse ecchymoses and blisters on a yellowish waxy base: a case of bullous amyloidosis. J Dermatol. 2016;43:713-714.
  13. Wang XD, Shen H, Liu ZH. Diffuse haemorrhagic bullous amyloidosis with multiple myeloma. Clin Exp Dermatol. 2008;33:94-96.
  14. Biswas P, Aggarwal I, Sen D, et al. Bullous pemphigoid clinically presenting as lichen amyloidosis. Indian J Dermatol Venereol Leprol. 2014;80:544-546.
  15. Bluhm JF 3rd. Bullous dermatosis vs amyloidosis. Arch Dermatol. 1981;117:252.
  16. Bluhm JF 3rd. Bullous amyloidosis vs epidermolysis bullosa acquisita. JAMA. 1981;245:32.
  17. Murphy GM, Wright J, Nicholls DS, et al. Sunbed-induced pseudoporphyria. Br J Dermatol. 1989;120:555-562.
  18. Pramatarov K, Lazarova A, Mateev G, et al. Bullous hemorrhagic primary systemic amyloidosis. Int J Dermatol. 1990;29:211-213.
  19. Bieber T, Ruzicka T, Linke RP, et al. Hemorrhagic bullous amyloidosis. a histologic, immunocytochemical, and ultrastructural study of two patients. Arch Dermatol. 1988;124:1683-1686.
  20. Khoo BP, Tay YK. Lichen amyloidosis: a bullous variant. Ann Acad Med Singapore. 2000;29:105-107.
  21. Asahina A, Hasegawa K, Ishiyama M, et al. Bullous amyloidosis mimicking bullous pemphigoid: usefulness of electron microscopic examination. Acta Derm Venereol. 2010;90:427-428.
  22. Schmutz JL, Barbaud A, Cuny JF, et al. Bullous amyloidosis [in French]. Ann Dermatol Venereol. 1988;115:295-301.
  23. Lachmann HJ, Hawkins PN. Amyloidosis of the skin. In: Goldsmith LA, Katz SI, Gilchrest BA, et al, eds. Fitzpatrick’s Dermatology in General Medicine. 8th ed. McGraw-Hill; 2012:1574-1583.
  24. Grundmann JU, Bonnekoh B, Gollnick H. Extensive haemorrhagic-bullous skin manifestation of systemic AA-amyloidosis associated with IgG lambda-myeloma. Eur J Dermatol. 2000;10:139-142.
  25. Hödl S, Turek TD, Kerl H. Plasmocytoma-associated bullous hemorrhagic amyloidosis of the skin [in German]. Hautarzt. 1982;33:556-558.
  26. Colucci G, Alberio L, Demarmels Biasiutti F, et al. Bilateral periorbital ecchymoses. an often missed sign of amyloid purpura. Hamostaseologie. 2014;34:249-252.
  27. Behera B, Pattnaik M, Sahu B, et al. Cutaneous manifestations of multiple myeloma. Indian J Dermatol. 2016;61:668-671.
  28. Fujita Y, Tsuji-Abe Y, Sato-Matsumura KC, et al. Nail dystrophy and blisters as sole manifestations in myeloma-associated amyloidosis. J Am Acad Dermatol. 2006;54:712-714.
  29. Chang SL, Lai PC, Cheng CJ, et al. Bullous amyloidosis in a hemodialysis patient is myeloma-associated rather than hemodialysis-associated amyloidosis. Amyloid. 2007;14:153-156.
  30. Winzer M, Ruppert M, Baretton G, et al. Bullous poikilodermatitic amyloidosis of the skin with junctional bulla development in IgG light chain plasmacytoma of the lambda type. histology, immunohistology and electron microscopy [in German]. Hautarzt. 1992;43:199-204.
References
  1. Weidner T, Illing T, Elsner P. Primary localized cutaneous amyloidosis: a systematic treatment review. Am J Clin Dermatol. 2017;18:629-642.
  2. Bolognia JL, Schaffer JV, Duncan KO, et al. Amyloidosis. Dermatology Essentials. Elsevier Saunders; 2014:341-345.
  3. Bhutani M, Shahid Z, Schnebelen A, et al. Cutaneous manifestations of multiple myeloma and other plasma cell proliferative disorders. Semin Oncol. 2016;43:395-400.
  4. Terushkin V, Boyd KP, Patel RR, et al. Primary localized cutaneous amyloidosis. Dermatol Online J. 2013;19:20711.
  5. LaChance A, Phelps A, Finch J, et al. Nodular localized primary cutaneous amyloidosis: a bullous variant. Clin Exp Dermatol. 2014;39:344-347.
  6. Gonzalez-Ramos J, Garrido-Gutiérrez C, González-Silva Y, et al. Relapsing bullous amyloidosis of the oral mucosa and acquired cutis laxa in a patient with multiple myeloma: a rare triple association. Clin Exp Dermatol. 2017;42:410-412.
  7. Kanoh T. Bullous amyloidosis [in Japanese]. Rinsho Ketsueki. 1993;34:1050-1052.
  8. Johnson TM, Rapini RP, Hebert AA, et al. Bullous amyloidosis. Cutis. 1989;43:346-352.
  9. Houman MH, Smiti KM, Ben Ghorbel I, et al. Bullous amyloidosis. Ann Dermatol Venereol. 2002;129:299-302.
  10. Sanusi T, Li Y, Qian Y, et al. Primary localized cutaneous nodular amyloidosis with bullous lesions. Indian J Dermatol Venereol Leprol. 2015;81:400-402.
  11. Ochiai T, Morishima T, Hao T, et al. Bullous amyloidosis: the mechanism of blister formation revealed by electron microscopy. J Cutan Pathol. 2001;28:407-411.
  12. Chu CH, Chan JY, Hsieh SW, et al. Diffuse ecchymoses and blisters on a yellowish waxy base: a case of bullous amyloidosis. J Dermatol. 2016;43:713-714.
  13. Wang XD, Shen H, Liu ZH. Diffuse haemorrhagic bullous amyloidosis with multiple myeloma. Clin Exp Dermatol. 2008;33:94-96.
  14. Biswas P, Aggarwal I, Sen D, et al. Bullous pemphigoid clinically presenting as lichen amyloidosis. Indian J Dermatol Venereol Leprol. 2014;80:544-546.
  15. Bluhm JF 3rd. Bullous dermatosis vs amyloidosis. Arch Dermatol. 1981;117:252.
  16. Bluhm JF 3rd. Bullous amyloidosis vs epidermolysis bullosa acquisita. JAMA. 1981;245:32.
  17. Murphy GM, Wright J, Nicholls DS, et al. Sunbed-induced pseudoporphyria. Br J Dermatol. 1989;120:555-562.
  18. Pramatarov K, Lazarova A, Mateev G, et al. Bullous hemorrhagic primary systemic amyloidosis. Int J Dermatol. 1990;29:211-213.
  19. Bieber T, Ruzicka T, Linke RP, et al. Hemorrhagic bullous amyloidosis. a histologic, immunocytochemical, and ultrastructural study of two patients. Arch Dermatol. 1988;124:1683-1686.
  20. Khoo BP, Tay YK. Lichen amyloidosis: a bullous variant. Ann Acad Med Singapore. 2000;29:105-107.
  21. Asahina A, Hasegawa K, Ishiyama M, et al. Bullous amyloidosis mimicking bullous pemphigoid: usefulness of electron microscopic examination. Acta Derm Venereol. 2010;90:427-428.
  22. Schmutz JL, Barbaud A, Cuny JF, et al. Bullous amyloidosis [in French]. Ann Dermatol Venereol. 1988;115:295-301.
  23. Lachmann HJ, Hawkins PN. Amyloidosis of the skin. In: Goldsmith LA, Katz SI, Gilchrest BA, et al, eds. Fitzpatrick’s Dermatology in General Medicine. 8th ed. McGraw-Hill; 2012:1574-1583.
  24. Grundmann JU, Bonnekoh B, Gollnick H. Extensive haemorrhagic-bullous skin manifestation of systemic AA-amyloidosis associated with IgG lambda-myeloma. Eur J Dermatol. 2000;10:139-142.
  25. Hödl S, Turek TD, Kerl H. Plasmocytoma-associated bullous hemorrhagic amyloidosis of the skin [in German]. Hautarzt. 1982;33:556-558.
  26. Colucci G, Alberio L, Demarmels Biasiutti F, et al. Bilateral periorbital ecchymoses. an often missed sign of amyloid purpura. Hamostaseologie. 2014;34:249-252.
  27. Behera B, Pattnaik M, Sahu B, et al. Cutaneous manifestations of multiple myeloma. Indian J Dermatol. 2016;61:668-671.
  28. Fujita Y, Tsuji-Abe Y, Sato-Matsumura KC, et al. Nail dystrophy and blisters as sole manifestations in myeloma-associated amyloidosis. J Am Acad Dermatol. 2006;54:712-714.
  29. Chang SL, Lai PC, Cheng CJ, et al. Bullous amyloidosis in a hemodialysis patient is myeloma-associated rather than hemodialysis-associated amyloidosis. Amyloid. 2007;14:153-156.
  30. Winzer M, Ruppert M, Baretton G, et al. Bullous poikilodermatitic amyloidosis of the skin with junctional bulla development in IgG light chain plasmacytoma of the lambda type. histology, immunohistology and electron microscopy [in German]. Hautarzt. 1992;43:199-204.
Issue
cutis - 108(3)
Issue
cutis - 108(3)
Page Number
e25-e28
Page Number
e25-e28
Publications
Publications
Topics
Article Type
Display Headline
Bullous Amyloidosis Masquerading as Pseudoporphyria
Display Headline
Bullous Amyloidosis Masquerading as Pseudoporphyria
Sections
Inside the Article

Practice Points

  • Primary systemic amyloidosis, including the rare cutaneous bullous amyloidosis, often is difficult to diagnose and has been associated with underlying plasma cell dyscrasia or multiple myeloma.
  • When evaluating patients with initially convincing signs of pseudoporphyria, it is imperative to consider the diagnosis of bullous amyloidosis, which additionally can present with intraoral hemorrhagic vesicles and have confirmatory histopathologic features.
  • Further investigation for multiple myeloma is warranted when patients with a chronic hemorrhagic bullous condition also present with symptoms of purpura, angina bullosa hemorrhagica, fatigue, weight loss, and/or macroglossia. Accurate diagnosis of bullous amyloidosis and timely treatment of its underlying cause will contribute to better, more proactive patient care.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media