LayerRx Mapping ID
508
Slot System
Featured Buckets
Featured Buckets Admin
Medscape Lead Concept
56

Cross-contamination of Pathology Specimens: A Cautionary Tale

Article Type
Changed
Thu, 01/10/2019 - 13:54
Display Headline
Cross-contamination of Pathology Specimens: A Cautionary Tale
In Collaboration with Cosmetic Surgery Forum

Cross-contamination of pathology specimens is a rare but nonnegligible source of potential morbidity in clinical practice. Contaminant tissue fragments, colloquially referred to as floaters, typically are readily identifiable based on obvious cytomorphologic differences, especially if the tissues arise from different organs; however, one cannot rely on such distinctions in a pathology laboratory dedicated to a single organ system (eg, dermatopathology). The inability to identify quickly and confidently the presence of a contaminant puts the patient at risk for misdiagnosis, which can lead to unnecessary morbidity or even mortality in the case of cancer misdiagnosis. Studies that have been conducted to estimate the incidence of this type of error have suggested an overall incidence rate between approximately 1% and 3%.1,2 Awareness of this phenomenon and careful scrutiny when the histopathologic evidence diverges considerably from the clinical impression is critical for minimizing the negative outcomes that could result from the presence of contaminant tissue. We present a case in which cross-contamination of a pathology specimen led to an initial erroneous diagnosis of an aggressive cutaneous melanoma in a patient with a benign adnexal neoplasm.

Case Report

A 72-year-old man was referred to the Pigmented Lesion and Melanoma Program at Stanford University Medical Center and Cancer Institute (Palo Alto, California) for evaluation and treatment of a presumed stage IIB melanoma on the right preauricular cheek based on a shave biopsy that had been performed (<1 month prior) by his local dermatology provider and subsequently read by an affiliated out-of-state dermatopathology laboratory. Per the clinical history that was gathered at the current presentation, neither the patient nor his wife had noticed the lesion prior to his dermatology provider pointing it out on the day of the biopsy. Additionally, he denied associated pain, bleeding, or ulceration. According to outside medical records, the referring dermatology provider described the lesion as a 4-mm pink pearly papule with telangiectasia favoring a diagnosis of basal cell carcinoma, and a diagnostic shave biopsy was performed. On presentation to our clinic, physical examination of the right preauricular cheek revealed a 4×3-mm depressed erythematous scar with no evidence of residual pigmentation or nodularity (Figure 1). There was no clinically appreciable regional lymphadenopathy.

Figure1
Figure 1. On physical examination at our clinic, a small pink scar (inner broken line) from a prior shave biopsy was noted on the patient’s right cheek. The outer broken line represents the proposed margins for wide local excision based on the initial diagnosis of a clinical stage IIB cutaneous melanoma.

The original dermatopathology report indicated an invasive melanoma with the following pathologic characteristics: superficial spreading type, Breslow depth of at least 2.16 mm, ulceration, and a mitotic index of 8 mitotic figures/mm2 with transection of the invasive component at the peripheral and deep margins. There was no evidence of regression, perineural invasion, lymphovascular invasion, or microsatellites. Interestingly, the report indicated that there also was a basaloid proliferation with features of cylindroma in the same pathology slide adjacent to the aggressive invasive melanoma that was described. Given the complexity of cases referred to our academic center, the standard of care includes internal dermatopathology review of all outside pathology specimens. This review proved critical to this patient’s care in light of the considerable divergence of the initial pathologic diagnosis and the reported clinical features of the lesion.

Internal review of the single pathology slide received from the referring provider showed a total of 4 sections, 3 of which are shown here (Figure 2A). Three sections, including the one not shown, were all consistent with a diagnosis of cylindroma and showed no evidence of a melanocytic proliferation (Figure 2B). However, the fourth section demonstrated marked morphologic dissimilarity compared to the other 3 sections. This outlier section showed a thick cutaneous melanoma with a Breslow depth of at least 2.1 mm, ulceration, a mitotic rate of 12 mitotic figures/mm2, and broad transection of the invasive component at the peripheral and deep margins (Figures 2C and 2D). Correlation with the gross description of tissue processing on the original pathology report indicating that the specimen had been trisected raised suspicion that the fourth and very dissimilar section could be a contaminant from another source that was incorporated into our patient’s histologic sections during processing. Taken together, these discrepancies made the diagnosis of cylindroma alone far more likely than cutaneous melanoma, but we needed conclusive evidence given the dramatic difference in prognosis and management between a cylindroma and an aggressive cutaneous melanoma.

Figure2
Figure 2. Upon review of 3 of 4 total sections on a single slide received from the dermatopathology laboratory where the specimen was processed, a malignant melanocytic neoplasm with epidermal ulceration was revealed (left), while 3 sections (middle and right as well as one not pictured due to image constraints) showed a benign basaloid neoplasm without epidermal ulceration (A)(H&E, original magnification ×2). On higher power, the middle section demonstrated a basaloid proliferation of well-differentiated cells in the dermis, which supported a diagnosis of cylindroma (B)(H&E, original magnification ×4), and the left section demonstrated a malignant melanocytic proliferation consisting of nested pleomorphic cells without maturation, which supported the diagnosis of invasive melanoma with ulceration (C)(H&E, original magnification ×4). Note the nested and pleomorphic characteristics of the densely packed melanocytes in the invasive melanoma (D)(H&E, original magnification ×20).

For further diagnostic clarification, we performed polymorphic short tandem repeat (STR) analysis, a well-described forensic pathology technique, to determine if the melanoma and cylindroma specimens derived from different patients, as we hypothesized. This analysis revealed differences in all but one DNA locus tested between the cylindroma specimen and the melanoma specimen, confirming our hypothesis (Figure 3). Subsequent discussion of the case with staff from the dermatopathology laboratory that processed this specimen provided further support for our suspicion that the invasive melanoma specimen was part of a case processed prior to our patient’s benign lesion. Therefore, the wide local excision for treatment of the suspected melanoma fortunately was canceled, and the patient did not require further treatment of the benign cylindroma. The patient expressed relief and gratitude for this critical clarification and change in management.

Figure3
Figure 3. Schematic representation of the principle on which short tandem repeat (STR) analysis for distinguishing one individual’s DNA from another is based.

 

 

Comment

Shah et al3 reported a similar case in which a benign granuloma of the lung masqueraded as a squamous cell carcinoma due to histopathologic contamination. Although few similar cases have been described in the literature, the risk posed by such contamination is remarkable, regardless of whether it occurs during specimen grossing, embedding, sectioning, or staining.1,4,5 This risk is amplified in facilities that process specimens originating predominantly from a single organ system or tissue type, as is often the case in dedicated dermatopathology laboratories. In this scenario, it is unlikely that one could use the presence of tissues from 2 different organ systems on a single slide as a way of easily recognizing the presence of a contaminant and rectifying the error. Additionally, the presence of malignant cells in the contaminant further complicates the problem and requires an investigation that can conclusively distinguish the contaminant from the patient’s actual tissue.

In our case, our dermatology and dermatopathology teams partnered with our molecular pathology team to find a solution. Polymorphic STR analysis via polymerase chain reaction amplification is a sensitive method employed commonly in forensic DNA laboratories for determining whether a sample submitted as evidence belongs to a given suspect.6 Although much more commonly used in forensics, STR analysis does have known roles in clinical medicine, such as chimerism testing after bone marrow or allogeneic stem cell transplantation.7 Given the relatively short period of time it takes along with the convenience of commercially available kits, a high discriminative ability, and well-validated interpretation procedures, STR analysis is an excellent method for determining if a given tissue sample came from a given patient, which is what was needed in our case.

The combined clinical, histopathologic, and molecular data in our case allowed for confident clarification of our patient’s diagnosis, sparing him the morbidity of wide local excision on the face, sentinel lymph node biopsy, and emotional distress associated with a diagnosis of aggressive cutaneous melanoma. Our case highlights the critical importance of internal review of pathology specimens in ensuring proper diagnosis and management and reminds us that, though rare, accidental contamination during processing of pathology specimens is a potential adverse event that must be considered, especially when a pathologic finding diverges considerably from what is anticipated based on the patient’s history and physical examination.

Acknowledgment
The authors express gratitude to the patient described herein who graciously provided permission for us to publish his case and clinical photography.

References
  1. Gephardt GN, Zarbo RJ. Extraneous tissue in surgical pathology: a College of American Pathologists Q-Probes study of 275 laboratories. Arch Pathol Lab Med. 1996;120:1009-1014.
  2. Alam M, Shah AD, Ali S, et al. Floaters in Mohs micrographic surgery [published online June 27, 2013]. Dermatol Surg. 2013;39:1317-1322.
  3. Shah PA, Prat MP, Hostler DC. Benign granuloma masquerading as squamous cell carcinoma due to a “floater.” Hawaii J Med Public Health. 2017;76(11, suppl 2):19-21.
  4. Platt E, Sommer P, McDonald L, et al. Tissue floaters and contaminants in the histology laboratory. Arch Pathol Lab Med. 2009;133:973-978.
  5. Layfield LJ, Witt BL, Metzger KG, et al. Extraneous tissue: a potential source for diagnostic error in surgical pathology. Am J Clin Pathol. 2011;136:767-772.
  6. Butler JM. Forensic DNA testing. Cold Spring Harb Protoc. 2011;2011:1438-1450.
  7. Manasatienkij C, Ra-ngabpai C. Clinical application of forensic DNA analysis: a literature review. J Med Assoc Thai. 2012;95:1357-1363.
Article PDF
Author and Disclosure Information

From Stanford Hospital and Clinics, Redwood City, California. Drs. Lewellis and Swetter are from the Department of Dermatology, Dr. Roy was from the Department of Pathology, Ms. Gojenola is from the Department of Pathology, and Dr. Rieger is from the Departments of Dermatology and Pathology. Dr. Roy currently is from the Dermatology Group of the Carolinas, Concord, North Carolina. Dr. Swetter also is from Dermatology Service, VA Palo Alto Health Care System, California.

The authors report no conflict of interest.

This study was part of a presentation at the 9th Cosmetic Surgery Forum under the direction of Joel Schlessinger, MD; November 29-December 2, 2017; Las Vegas, Nevada. Dr. Lewellis was a Top 10 Fellow and Resident Grant winner.

Correspondence: Stephen W. Lewellis, MD, PhD, Department of Dermatology, Stanford Hospital and Clinics, 450 Broadway St, Pavilion B, 4th Floor, Redwood City, CA 94063 (lewellis@standford.edu).

Issue
Cutis - 102(4)
Publications
Topics
Page Number
E12-E14
Sections
Author and Disclosure Information

From Stanford Hospital and Clinics, Redwood City, California. Drs. Lewellis and Swetter are from the Department of Dermatology, Dr. Roy was from the Department of Pathology, Ms. Gojenola is from the Department of Pathology, and Dr. Rieger is from the Departments of Dermatology and Pathology. Dr. Roy currently is from the Dermatology Group of the Carolinas, Concord, North Carolina. Dr. Swetter also is from Dermatology Service, VA Palo Alto Health Care System, California.

The authors report no conflict of interest.

This study was part of a presentation at the 9th Cosmetic Surgery Forum under the direction of Joel Schlessinger, MD; November 29-December 2, 2017; Las Vegas, Nevada. Dr. Lewellis was a Top 10 Fellow and Resident Grant winner.

Correspondence: Stephen W. Lewellis, MD, PhD, Department of Dermatology, Stanford Hospital and Clinics, 450 Broadway St, Pavilion B, 4th Floor, Redwood City, CA 94063 (lewellis@standford.edu).

Author and Disclosure Information

From Stanford Hospital and Clinics, Redwood City, California. Drs. Lewellis and Swetter are from the Department of Dermatology, Dr. Roy was from the Department of Pathology, Ms. Gojenola is from the Department of Pathology, and Dr. Rieger is from the Departments of Dermatology and Pathology. Dr. Roy currently is from the Dermatology Group of the Carolinas, Concord, North Carolina. Dr. Swetter also is from Dermatology Service, VA Palo Alto Health Care System, California.

The authors report no conflict of interest.

This study was part of a presentation at the 9th Cosmetic Surgery Forum under the direction of Joel Schlessinger, MD; November 29-December 2, 2017; Las Vegas, Nevada. Dr. Lewellis was a Top 10 Fellow and Resident Grant winner.

Correspondence: Stephen W. Lewellis, MD, PhD, Department of Dermatology, Stanford Hospital and Clinics, 450 Broadway St, Pavilion B, 4th Floor, Redwood City, CA 94063 (lewellis@standford.edu).

Article PDF
Article PDF
In Collaboration with Cosmetic Surgery Forum
In Collaboration with Cosmetic Surgery Forum

Cross-contamination of pathology specimens is a rare but nonnegligible source of potential morbidity in clinical practice. Contaminant tissue fragments, colloquially referred to as floaters, typically are readily identifiable based on obvious cytomorphologic differences, especially if the tissues arise from different organs; however, one cannot rely on such distinctions in a pathology laboratory dedicated to a single organ system (eg, dermatopathology). The inability to identify quickly and confidently the presence of a contaminant puts the patient at risk for misdiagnosis, which can lead to unnecessary morbidity or even mortality in the case of cancer misdiagnosis. Studies that have been conducted to estimate the incidence of this type of error have suggested an overall incidence rate between approximately 1% and 3%.1,2 Awareness of this phenomenon and careful scrutiny when the histopathologic evidence diverges considerably from the clinical impression is critical for minimizing the negative outcomes that could result from the presence of contaminant tissue. We present a case in which cross-contamination of a pathology specimen led to an initial erroneous diagnosis of an aggressive cutaneous melanoma in a patient with a benign adnexal neoplasm.

Case Report

A 72-year-old man was referred to the Pigmented Lesion and Melanoma Program at Stanford University Medical Center and Cancer Institute (Palo Alto, California) for evaluation and treatment of a presumed stage IIB melanoma on the right preauricular cheek based on a shave biopsy that had been performed (<1 month prior) by his local dermatology provider and subsequently read by an affiliated out-of-state dermatopathology laboratory. Per the clinical history that was gathered at the current presentation, neither the patient nor his wife had noticed the lesion prior to his dermatology provider pointing it out on the day of the biopsy. Additionally, he denied associated pain, bleeding, or ulceration. According to outside medical records, the referring dermatology provider described the lesion as a 4-mm pink pearly papule with telangiectasia favoring a diagnosis of basal cell carcinoma, and a diagnostic shave biopsy was performed. On presentation to our clinic, physical examination of the right preauricular cheek revealed a 4×3-mm depressed erythematous scar with no evidence of residual pigmentation or nodularity (Figure 1). There was no clinically appreciable regional lymphadenopathy.

Figure1
Figure 1. On physical examination at our clinic, a small pink scar (inner broken line) from a prior shave biopsy was noted on the patient’s right cheek. The outer broken line represents the proposed margins for wide local excision based on the initial diagnosis of a clinical stage IIB cutaneous melanoma.

The original dermatopathology report indicated an invasive melanoma with the following pathologic characteristics: superficial spreading type, Breslow depth of at least 2.16 mm, ulceration, and a mitotic index of 8 mitotic figures/mm2 with transection of the invasive component at the peripheral and deep margins. There was no evidence of regression, perineural invasion, lymphovascular invasion, or microsatellites. Interestingly, the report indicated that there also was a basaloid proliferation with features of cylindroma in the same pathology slide adjacent to the aggressive invasive melanoma that was described. Given the complexity of cases referred to our academic center, the standard of care includes internal dermatopathology review of all outside pathology specimens. This review proved critical to this patient’s care in light of the considerable divergence of the initial pathologic diagnosis and the reported clinical features of the lesion.

Internal review of the single pathology slide received from the referring provider showed a total of 4 sections, 3 of which are shown here (Figure 2A). Three sections, including the one not shown, were all consistent with a diagnosis of cylindroma and showed no evidence of a melanocytic proliferation (Figure 2B). However, the fourth section demonstrated marked morphologic dissimilarity compared to the other 3 sections. This outlier section showed a thick cutaneous melanoma with a Breslow depth of at least 2.1 mm, ulceration, a mitotic rate of 12 mitotic figures/mm2, and broad transection of the invasive component at the peripheral and deep margins (Figures 2C and 2D). Correlation with the gross description of tissue processing on the original pathology report indicating that the specimen had been trisected raised suspicion that the fourth and very dissimilar section could be a contaminant from another source that was incorporated into our patient’s histologic sections during processing. Taken together, these discrepancies made the diagnosis of cylindroma alone far more likely than cutaneous melanoma, but we needed conclusive evidence given the dramatic difference in prognosis and management between a cylindroma and an aggressive cutaneous melanoma.

Figure2
Figure 2. Upon review of 3 of 4 total sections on a single slide received from the dermatopathology laboratory where the specimen was processed, a malignant melanocytic neoplasm with epidermal ulceration was revealed (left), while 3 sections (middle and right as well as one not pictured due to image constraints) showed a benign basaloid neoplasm without epidermal ulceration (A)(H&E, original magnification ×2). On higher power, the middle section demonstrated a basaloid proliferation of well-differentiated cells in the dermis, which supported a diagnosis of cylindroma (B)(H&E, original magnification ×4), and the left section demonstrated a malignant melanocytic proliferation consisting of nested pleomorphic cells without maturation, which supported the diagnosis of invasive melanoma with ulceration (C)(H&E, original magnification ×4). Note the nested and pleomorphic characteristics of the densely packed melanocytes in the invasive melanoma (D)(H&E, original magnification ×20).

For further diagnostic clarification, we performed polymorphic short tandem repeat (STR) analysis, a well-described forensic pathology technique, to determine if the melanoma and cylindroma specimens derived from different patients, as we hypothesized. This analysis revealed differences in all but one DNA locus tested between the cylindroma specimen and the melanoma specimen, confirming our hypothesis (Figure 3). Subsequent discussion of the case with staff from the dermatopathology laboratory that processed this specimen provided further support for our suspicion that the invasive melanoma specimen was part of a case processed prior to our patient’s benign lesion. Therefore, the wide local excision for treatment of the suspected melanoma fortunately was canceled, and the patient did not require further treatment of the benign cylindroma. The patient expressed relief and gratitude for this critical clarification and change in management.

Figure3
Figure 3. Schematic representation of the principle on which short tandem repeat (STR) analysis for distinguishing one individual’s DNA from another is based.

 

 

Comment

Shah et al3 reported a similar case in which a benign granuloma of the lung masqueraded as a squamous cell carcinoma due to histopathologic contamination. Although few similar cases have been described in the literature, the risk posed by such contamination is remarkable, regardless of whether it occurs during specimen grossing, embedding, sectioning, or staining.1,4,5 This risk is amplified in facilities that process specimens originating predominantly from a single organ system or tissue type, as is often the case in dedicated dermatopathology laboratories. In this scenario, it is unlikely that one could use the presence of tissues from 2 different organ systems on a single slide as a way of easily recognizing the presence of a contaminant and rectifying the error. Additionally, the presence of malignant cells in the contaminant further complicates the problem and requires an investigation that can conclusively distinguish the contaminant from the patient’s actual tissue.

In our case, our dermatology and dermatopathology teams partnered with our molecular pathology team to find a solution. Polymorphic STR analysis via polymerase chain reaction amplification is a sensitive method employed commonly in forensic DNA laboratories for determining whether a sample submitted as evidence belongs to a given suspect.6 Although much more commonly used in forensics, STR analysis does have known roles in clinical medicine, such as chimerism testing after bone marrow or allogeneic stem cell transplantation.7 Given the relatively short period of time it takes along with the convenience of commercially available kits, a high discriminative ability, and well-validated interpretation procedures, STR analysis is an excellent method for determining if a given tissue sample came from a given patient, which is what was needed in our case.

The combined clinical, histopathologic, and molecular data in our case allowed for confident clarification of our patient’s diagnosis, sparing him the morbidity of wide local excision on the face, sentinel lymph node biopsy, and emotional distress associated with a diagnosis of aggressive cutaneous melanoma. Our case highlights the critical importance of internal review of pathology specimens in ensuring proper diagnosis and management and reminds us that, though rare, accidental contamination during processing of pathology specimens is a potential adverse event that must be considered, especially when a pathologic finding diverges considerably from what is anticipated based on the patient’s history and physical examination.

Acknowledgment
The authors express gratitude to the patient described herein who graciously provided permission for us to publish his case and clinical photography.

Cross-contamination of pathology specimens is a rare but nonnegligible source of potential morbidity in clinical practice. Contaminant tissue fragments, colloquially referred to as floaters, typically are readily identifiable based on obvious cytomorphologic differences, especially if the tissues arise from different organs; however, one cannot rely on such distinctions in a pathology laboratory dedicated to a single organ system (eg, dermatopathology). The inability to identify quickly and confidently the presence of a contaminant puts the patient at risk for misdiagnosis, which can lead to unnecessary morbidity or even mortality in the case of cancer misdiagnosis. Studies that have been conducted to estimate the incidence of this type of error have suggested an overall incidence rate between approximately 1% and 3%.1,2 Awareness of this phenomenon and careful scrutiny when the histopathologic evidence diverges considerably from the clinical impression is critical for minimizing the negative outcomes that could result from the presence of contaminant tissue. We present a case in which cross-contamination of a pathology specimen led to an initial erroneous diagnosis of an aggressive cutaneous melanoma in a patient with a benign adnexal neoplasm.

Case Report

A 72-year-old man was referred to the Pigmented Lesion and Melanoma Program at Stanford University Medical Center and Cancer Institute (Palo Alto, California) for evaluation and treatment of a presumed stage IIB melanoma on the right preauricular cheek based on a shave biopsy that had been performed (<1 month prior) by his local dermatology provider and subsequently read by an affiliated out-of-state dermatopathology laboratory. Per the clinical history that was gathered at the current presentation, neither the patient nor his wife had noticed the lesion prior to his dermatology provider pointing it out on the day of the biopsy. Additionally, he denied associated pain, bleeding, or ulceration. According to outside medical records, the referring dermatology provider described the lesion as a 4-mm pink pearly papule with telangiectasia favoring a diagnosis of basal cell carcinoma, and a diagnostic shave biopsy was performed. On presentation to our clinic, physical examination of the right preauricular cheek revealed a 4×3-mm depressed erythematous scar with no evidence of residual pigmentation or nodularity (Figure 1). There was no clinically appreciable regional lymphadenopathy.

Figure1
Figure 1. On physical examination at our clinic, a small pink scar (inner broken line) from a prior shave biopsy was noted on the patient’s right cheek. The outer broken line represents the proposed margins for wide local excision based on the initial diagnosis of a clinical stage IIB cutaneous melanoma.

The original dermatopathology report indicated an invasive melanoma with the following pathologic characteristics: superficial spreading type, Breslow depth of at least 2.16 mm, ulceration, and a mitotic index of 8 mitotic figures/mm2 with transection of the invasive component at the peripheral and deep margins. There was no evidence of regression, perineural invasion, lymphovascular invasion, or microsatellites. Interestingly, the report indicated that there also was a basaloid proliferation with features of cylindroma in the same pathology slide adjacent to the aggressive invasive melanoma that was described. Given the complexity of cases referred to our academic center, the standard of care includes internal dermatopathology review of all outside pathology specimens. This review proved critical to this patient’s care in light of the considerable divergence of the initial pathologic diagnosis and the reported clinical features of the lesion.

Internal review of the single pathology slide received from the referring provider showed a total of 4 sections, 3 of which are shown here (Figure 2A). Three sections, including the one not shown, were all consistent with a diagnosis of cylindroma and showed no evidence of a melanocytic proliferation (Figure 2B). However, the fourth section demonstrated marked morphologic dissimilarity compared to the other 3 sections. This outlier section showed a thick cutaneous melanoma with a Breslow depth of at least 2.1 mm, ulceration, a mitotic rate of 12 mitotic figures/mm2, and broad transection of the invasive component at the peripheral and deep margins (Figures 2C and 2D). Correlation with the gross description of tissue processing on the original pathology report indicating that the specimen had been trisected raised suspicion that the fourth and very dissimilar section could be a contaminant from another source that was incorporated into our patient’s histologic sections during processing. Taken together, these discrepancies made the diagnosis of cylindroma alone far more likely than cutaneous melanoma, but we needed conclusive evidence given the dramatic difference in prognosis and management between a cylindroma and an aggressive cutaneous melanoma.

Figure2
Figure 2. Upon review of 3 of 4 total sections on a single slide received from the dermatopathology laboratory where the specimen was processed, a malignant melanocytic neoplasm with epidermal ulceration was revealed (left), while 3 sections (middle and right as well as one not pictured due to image constraints) showed a benign basaloid neoplasm without epidermal ulceration (A)(H&E, original magnification ×2). On higher power, the middle section demonstrated a basaloid proliferation of well-differentiated cells in the dermis, which supported a diagnosis of cylindroma (B)(H&E, original magnification ×4), and the left section demonstrated a malignant melanocytic proliferation consisting of nested pleomorphic cells without maturation, which supported the diagnosis of invasive melanoma with ulceration (C)(H&E, original magnification ×4). Note the nested and pleomorphic characteristics of the densely packed melanocytes in the invasive melanoma (D)(H&E, original magnification ×20).

For further diagnostic clarification, we performed polymorphic short tandem repeat (STR) analysis, a well-described forensic pathology technique, to determine if the melanoma and cylindroma specimens derived from different patients, as we hypothesized. This analysis revealed differences in all but one DNA locus tested between the cylindroma specimen and the melanoma specimen, confirming our hypothesis (Figure 3). Subsequent discussion of the case with staff from the dermatopathology laboratory that processed this specimen provided further support for our suspicion that the invasive melanoma specimen was part of a case processed prior to our patient’s benign lesion. Therefore, the wide local excision for treatment of the suspected melanoma fortunately was canceled, and the patient did not require further treatment of the benign cylindroma. The patient expressed relief and gratitude for this critical clarification and change in management.

Figure3
Figure 3. Schematic representation of the principle on which short tandem repeat (STR) analysis for distinguishing one individual’s DNA from another is based.

 

 

Comment

Shah et al3 reported a similar case in which a benign granuloma of the lung masqueraded as a squamous cell carcinoma due to histopathologic contamination. Although few similar cases have been described in the literature, the risk posed by such contamination is remarkable, regardless of whether it occurs during specimen grossing, embedding, sectioning, or staining.1,4,5 This risk is amplified in facilities that process specimens originating predominantly from a single organ system or tissue type, as is often the case in dedicated dermatopathology laboratories. In this scenario, it is unlikely that one could use the presence of tissues from 2 different organ systems on a single slide as a way of easily recognizing the presence of a contaminant and rectifying the error. Additionally, the presence of malignant cells in the contaminant further complicates the problem and requires an investigation that can conclusively distinguish the contaminant from the patient’s actual tissue.

In our case, our dermatology and dermatopathology teams partnered with our molecular pathology team to find a solution. Polymorphic STR analysis via polymerase chain reaction amplification is a sensitive method employed commonly in forensic DNA laboratories for determining whether a sample submitted as evidence belongs to a given suspect.6 Although much more commonly used in forensics, STR analysis does have known roles in clinical medicine, such as chimerism testing after bone marrow or allogeneic stem cell transplantation.7 Given the relatively short period of time it takes along with the convenience of commercially available kits, a high discriminative ability, and well-validated interpretation procedures, STR analysis is an excellent method for determining if a given tissue sample came from a given patient, which is what was needed in our case.

The combined clinical, histopathologic, and molecular data in our case allowed for confident clarification of our patient’s diagnosis, sparing him the morbidity of wide local excision on the face, sentinel lymph node biopsy, and emotional distress associated with a diagnosis of aggressive cutaneous melanoma. Our case highlights the critical importance of internal review of pathology specimens in ensuring proper diagnosis and management and reminds us that, though rare, accidental contamination during processing of pathology specimens is a potential adverse event that must be considered, especially when a pathologic finding diverges considerably from what is anticipated based on the patient’s history and physical examination.

Acknowledgment
The authors express gratitude to the patient described herein who graciously provided permission for us to publish his case and clinical photography.

References
  1. Gephardt GN, Zarbo RJ. Extraneous tissue in surgical pathology: a College of American Pathologists Q-Probes study of 275 laboratories. Arch Pathol Lab Med. 1996;120:1009-1014.
  2. Alam M, Shah AD, Ali S, et al. Floaters in Mohs micrographic surgery [published online June 27, 2013]. Dermatol Surg. 2013;39:1317-1322.
  3. Shah PA, Prat MP, Hostler DC. Benign granuloma masquerading as squamous cell carcinoma due to a “floater.” Hawaii J Med Public Health. 2017;76(11, suppl 2):19-21.
  4. Platt E, Sommer P, McDonald L, et al. Tissue floaters and contaminants in the histology laboratory. Arch Pathol Lab Med. 2009;133:973-978.
  5. Layfield LJ, Witt BL, Metzger KG, et al. Extraneous tissue: a potential source for diagnostic error in surgical pathology. Am J Clin Pathol. 2011;136:767-772.
  6. Butler JM. Forensic DNA testing. Cold Spring Harb Protoc. 2011;2011:1438-1450.
  7. Manasatienkij C, Ra-ngabpai C. Clinical application of forensic DNA analysis: a literature review. J Med Assoc Thai. 2012;95:1357-1363.
References
  1. Gephardt GN, Zarbo RJ. Extraneous tissue in surgical pathology: a College of American Pathologists Q-Probes study of 275 laboratories. Arch Pathol Lab Med. 1996;120:1009-1014.
  2. Alam M, Shah AD, Ali S, et al. Floaters in Mohs micrographic surgery [published online June 27, 2013]. Dermatol Surg. 2013;39:1317-1322.
  3. Shah PA, Prat MP, Hostler DC. Benign granuloma masquerading as squamous cell carcinoma due to a “floater.” Hawaii J Med Public Health. 2017;76(11, suppl 2):19-21.
  4. Platt E, Sommer P, McDonald L, et al. Tissue floaters and contaminants in the histology laboratory. Arch Pathol Lab Med. 2009;133:973-978.
  5. Layfield LJ, Witt BL, Metzger KG, et al. Extraneous tissue: a potential source for diagnostic error in surgical pathology. Am J Clin Pathol. 2011;136:767-772.
  6. Butler JM. Forensic DNA testing. Cold Spring Harb Protoc. 2011;2011:1438-1450.
  7. Manasatienkij C, Ra-ngabpai C. Clinical application of forensic DNA analysis: a literature review. J Med Assoc Thai. 2012;95:1357-1363.
Issue
Cutis - 102(4)
Issue
Cutis - 102(4)
Page Number
E12-E14
Page Number
E12-E14
Publications
Publications
Topics
Article Type
Display Headline
Cross-contamination of Pathology Specimens: A Cautionary Tale
Display Headline
Cross-contamination of Pathology Specimens: A Cautionary Tale
Sections
Inside the Article

Resident Pearl

  • Although cross-contamination of pathology specimens is rare, it does occur and can impact diagnosis and management if detected early.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Article PDF Media

Cutaneous Angiosarcoma of the Lower Leg

Article Type
Changed
Thu, 01/10/2019 - 13:54
Display Headline
Cutaneous Angiosarcoma of the Lower Leg

Angiosarcoma is a rare and aggressive vascular malignant neoplasm derived from endothelial cells. In general, sarcomas account for approximately 1% of all malignancies, with approximately 2% being angiosarcomas.1 The risk of recurrence at 5 years is estimated to be 84%, and 5-year survival is estimated at 15% to 30%. Poor prognostic factors for angiosarcoma include large tumor size, depth of invasion greater than 3 mm, high mitotic rate, positive surgical margins, and metastasis.2 Approximately 20% to 40% of patients who are diagnosed with angiosarcoma already have distant metastasis, contributing to the aggressive nature of this neoplasm.3

Angiosarcoma can affect various anatomic locations, including the skin, soft tissue, breasts, and liver. Cutaneous angiosarcoma is the most common clinical manifestation, accounting for approximately 50% to 60% of all cases, and typically is known to occur in 3 distinct settings.2 Primary or idiopathic cutaneous angiosarcoma is most commonly seen in elderly individuals, with a peak incidence in the seventh to eighth decades of life, and presents as a bruiselike lesion predominantly on the head and neck. Angiosarcoma also is seen clinically in patients exposed to radiation treatment, with a median onset of symptoms occurring 5 to 10 years posttreatment, and in patients with chronic lymphedema, usually on the arm following radical mastectomy, which also is known as Stewart-Treves syndrome.2

With any sarcoma, treatment typically first involves surgical excision; however, there is no direct approach for treatment of cutaneous angiosarcoma, as an individual plan typically is needed for each patient. Treatment options include surgical excision, radiation, chemotherapy, or a combination of these therapies.2,4

We present a rare case of cutaneous angiosarcoma of the left leg in the setting of chronic venous insufficiency with some degree of lymphedema and a nonhealing ulcer. This case is unique in that it does not fit the classic presentation of cutaneous angiosarcoma previously described.

Case Report

An 83-year-old woman with a medical history of advanced dementia, congestive heart failure, chronic obstructive pulmonary disease, chronic kidney disease, type 2 diabetes mellitus, hypertension, and chronic venous insufficiency with stasis dermatitis presented to the emergency department following a mechanical fall. Most of her medical history was obtained from the patient’s family. She had a history of multiple falls originally thought to be related to a chronic leg ulcer that had been managed with wound care. Recently, however, the lesion was noted to have increasing erythema surrounding the wound margins. An 8×8-cm erythematous plaque on the anterior lateral left leg with a firm central nodule with hemorrhagic crust that measured approximately 4 cm in diameter was noted by the emergency department physicians (Figure 1). In the emergency department, vitals and other laboratory values were within reference range, and a radiograph of the left tibia/fibula was unremarkable. Cellulitis initially was considered in the emergency department and cephalexin was started; however, since the patient was afebrile and had no leukocytosis, plastic surgery also was consulted. Biopsies were obtained from the superior and inferior parts of the lesion. Histologic analysis revealed a poorly differentiated vascular neoplasm of epithelioid endothelial cells with considerable cell atypia that extended through the entirety of the dermis (Figure 2). The tumor cells stained positive with vimentin and CD34. Pathology noted no immunohistochemistry stains to synaptophysin, S-100, human melanoma black 45, MART-1, CK20, CK7, CK8/18, CK5/6, and p63. The pathologic diagnosis was consistent with cutaneous angiosarcoma. Computed tomography of the chest, abdomen, and pelvis revealed no local or distant metastases.

Figure1
Figure 1. Cutaneous angiosarcoma presenting as a large erythematous plaque on the anterior lateral left lower leg with a firm central nodule with overlying hemorrhagic crust.

Figure2
Figure 2. Histologic analysis revealed a poorly differentiated vascular neoplasm extending through the dermis (A) with epithelioid endothelial cells (B)(H&E, original magnifications ×40 and ×100). Considerable cell atypia and mitotic figures were appreciated on higher power (C)(H&E, original magnification ×400).

A wide excision of the cutaneous angiosarcoma was performed. The initial frozen section analysis revealed positive margins. Three additional excisions still showed positive margins, and further excision was held after obtaining family consent due to the extensive nature of the neoplasm and lengthy operating room time. The final defect after excision measured 15×10×2.5 cm (Figure 3A), and subsequent application of a split-thickness graft was performed. Additional treatment options were discussed with the family, including radiation therapy, amputation of the left lower leg, or no treatment. The family opted not to proceed with further treatment. The graft healed without signs of reoccurrence approximately 3 months later (Figure 3B), and the patient received physical therapy, which allowed her to gain strength and some independence.

Figure3
Figure 3. Wide surgical excision of the cutaneous angiosarcoma yielded a final defect measuring 15×10×2.5 cm (A). Approximately 3 months following excision and subsequent split-thickness skin graft, the patient was healing well with no evidence of reoccurrence (B).

 

 

Comment

Clinical Manifestation
Cutaneous angiosarcoma is a rare malignant vascular neoplasm that when clinically diagnosed is typically seen in 3 settings: (1) idiopathic (commonly on the face and neck), (2) following radiation treatment, and (3) classically following mastectomy with subsequent chronic lymphedema. Our patient did not classically fit these settings of cutaneous angiosarcoma due to the location of the lesion on the lower leg as well as its occurrence in the setting of a chronic nonhealing ulcer and lymphedema.

Chronic lymphedema is a common clinical manifestation that is likely secondary to other medical conditions, such as in our patient. As a result, these patients are at increased risk for developing chronic ulcers due to poor wound healing; however, as seen in our patient, chronic nonhealing ulcers require a broad differential because they may clinically mimic many processes. Patient history and visual presentation were crucial in this case because a biopsy was obtained that ultimately led to the patient’s diagnosis.

Differential Diagnosis
Initially, a venous ulcer secondary to chronic venous insufficiency was considered in the differential for our patient. She had a history of congestive heart failure, kidney disease, and type 2 diabetes mellitus, all of which contribute to lymphedema and/or poor wound healing. However, venous ulcers usually are located on the medial ankles and are irregularly shaped with an erythematous border and fibrinous exudate with central depression, making it a less likely diagnosis in our patient. Additionally, an infectious process was considered, but the patient was afebrile and laboratory values demonstrated no leukocytosis.

Marjolin ulcer was highly suspected because the clinical presentation revealed a nodule with hemorrhagic crust and induration in the setting of a chronic nonhealing ulcer. The pathogenesis of malignancy in chronic ulcers is thought to be due to continuous mitotic activity from regeneration and repair of the wound, especially in the setting of repeated trauma to the area.5 In our patient, the history of multiple falls with possible multitrauma injury to the chronic ulcer further increased suspicion of malignancy. The most common and frequently seen malignancy that develops in chronic ulcers is squamous cell carcinoma (SCC) followed by basal cell carcinoma. Plastic surgery suspected an SCC for the working diagnosis, which prompted a punch biopsy; however, the histologic analysis was not consistent with SCC or basal cell carcinoma. Marjolin ulcer also may demonstrate a periosteal reaction,5 which was not the case with our patient after a radiograph of the left tibia/fibula was unremarkable.

Another potential malignancy to consider is melanoma. There are few case reports of biopsy-proven melanoma from an enlarging chronic ulcer.6,7 Additionally, poorly differentiated angiosarcoma can mimic melanoma2; however, immunohistochemistry stain was negative for S-100, human melanoma black 45, and MART-1, making melanoma unlikely.

Kaposi sarcoma (KS) and angiosarcoma are both malignant vascular tumors that similarly present with red to purple patches, plaques, or nodules, making it difficult to distinguish between the two conditions. It is important to note that KS usually is lower grade, and the pathogenesis is linked to human herpesvirus 8, which can be identified on immunohistochemistry staining. There have been cases of KS reported in patients who have no history of human immunodeficiency virus/AIDS, thus the classic subtype of KS may have been considered in this patient.8 The histologic appearance of KS may vary from dilated irregular endothelial cells lining the vascular space to mild endothelial cell atypia. Histology also shows hemosiderin-laden macrophages, extravasated red blood cells, and an inflammatory infiltrate. An additional malignant vascular neoplasm that needs to be differentiated is epithelioid hemangioendothelioma. Cutaneous presentation of an epithelioid hemangioendothelioma may be similar to what was seen in our patient but histologically will usually show neoplastic cells with pale eosinophilic cytoplasm and vesicular nuclei of plump, oval, polygonal cells in cords or aggregates surrounding vascular channels. These neoplasms also tend to occur around medium- to large-sized veins.1,9 With our patient, even though human herpesvirus 8 was not tested with immunohistochemistry, gold standard immunohistochemistry confirmation with CD34 and vimentin staining combined with poorly differentiated endothelial atypia with mitotic figures on histologic analysis favored angiosarcoma versus KS or epithelioid hemangioendothelioma.10,11

Management
Cutaneous angiosarcoma is a rare and aggressive vascular neoplasm accounting for approximately 2% of all combined sarcomas, with an estimated 20% to 40% having distant metastasis at diagnosis.1,3 For this reason, computed tomography was performed in our patient and revealed no local or distant metastasis. Therefore, chemotherapy was not an appropriate adjuvant treatment option.12 With no evidence of metastasis, initial treatment began with surgical removal but proved to be difficult in our patient. Although the implications of positive surgical margins remain unclear with regard to overall patient survival, surgical resection followed by radiation therapy has been shown to be optimal, as it reduces the risk of local reoccurrence.3 There have been reported cases of cutaneous angiosarcoma of the leg that were treated with amputation without signs of reoccurrence or metastasis.10,13,14 Given the results from these cases and considering that our patient had no metastasis, amputation seemed to be a good prognostic option; however, considering other factors regarding the patient’s comorbidities and quality of life, her family decided not to pursue any further treatment with amputation or radiation therapy.

Conclusion

There should be low threshold for biopsy in patients who present with nonhealing wounds that do not progress in the normal phase of wound healing with suspicion for malignancy. As seen with our patient, cutaneous angiosarcoma can clinically mimic many disease processes, and although rare in nature, it should always be considered when a patient presents with a rapidly growing lesion in the setting of chronic lymphedema or venous ulcer.

References
  1. Kumar V, Abbas A, Aster J. Robbins Basic Pathology. 9th ed. Philadelphia, PA: Elsevier Saunders; 2013.
  2. Donghi D, Kerl K, Dummer R, et al. Cutaneous angiosarcoma: own experience over 13 years. clinical features, disease course and immunohistochemical profile. J Eur Acad Dermatol Venereol. 2010;24:1230-1234.
  3. Dossett LA, Harrington M, Cruse CW, et al. Cutaneous angiosarcoma. Curr Probl Cancer. 2015;39:258-263.
  4. Morgan MB, Swann M, Somach S, et al. Cutaneous angiosarcoma: a case series with prognostic correlation. J Am Acad Dermatol. 2004;50:867-874.
  5. Pekarek B, Buck S, Osher L. A comprehensive review on Marjolin’s ulcers: diagnosis and treatment. J Am Col Certif Wound Spec. 2011;3:60-64.
  6. Gerslova A, Pokorna A, Stukavcova A, et al. Rare cause of non-healing foot wound—acral lentiginous melanoma. Neuro Endocrinol Lett. 2012;37:12-17.
  7. Turk BG, Bozkurt A, Yaman B, et al. Melanoma arising in chronic ulceration associated with lymphoedema. J Wound Care. 2013;22:74-75.
  8. Phavixay L, Raynolds D, Simman R. Non AIDS Kaposi’s sarcoma leading to lower extremities wounds, case presentations and discussion.J Am Coll Clin Wound Spec. 2012;4:13-15.
  9. Requena L, Kutzner H. Hemangioendothelioma. Semin Diagn Pathol. 2013;30:29-44.
  10. Harrison WD, Chandrasekar CR. Stewart-Treves syndrome following idiopathic leg lymphoedema: remember sarcoma. J Wound Care. 2015;24(6 suppl):S5-S7.
  11. Kak I, Salama S, Gohla G, et al. A case of patch stage of Kaposi’s sarcoma and discussion of the differential diagnosis. Rare Tumors. 2016;8:6123.
  12. Agulnik M, Yarber JL, Okuno SH, et al. An open-label, multicenter, phase II study of bevacizumab for the treatment of angiosarcoma and epithelioid hemangioendotheliomas. Ann Oncol. 2013;24:257-263.
  13. Linda DD, Harish S, Alowami S, et al. Radiology-pathology conference: cutaneous angiosarcoma of the leg. Clin Imaging. 2013;37:602-607.
  14. Roy P, Clark MA, Thomas JM. Stewart-Treves syndrome—treatment and outcome in six patients from a single centre. Eur J Surg Oncol. 2004;30:982-986.
Article PDF
Author and Disclosure Information

Dr. Scholtz is from the Department of Dermatology, Wright State University, Dayton, Ohio. Dr. Mishra is from the Department of Pathology, Trillium Pathology Inc, Springfield, Ohio. Dr. Simman is from the Wright State University Boonshoft School of Medicine, Dayton, and Jobst Vascular Institute/ProMedica Health System Toledo Hospital, Ohio.

The authors report no conflict of interest.

Correspondence: Jaclyn Scholtz, MD, Department of Dermatology, 725 University Blvd, Dayton, OH 45435 (scholtz.2@wright.edu).

Issue
Cutis - 102(4)
Publications
Topics
Page Number
E8-E11
Sections
Author and Disclosure Information

Dr. Scholtz is from the Department of Dermatology, Wright State University, Dayton, Ohio. Dr. Mishra is from the Department of Pathology, Trillium Pathology Inc, Springfield, Ohio. Dr. Simman is from the Wright State University Boonshoft School of Medicine, Dayton, and Jobst Vascular Institute/ProMedica Health System Toledo Hospital, Ohio.

The authors report no conflict of interest.

Correspondence: Jaclyn Scholtz, MD, Department of Dermatology, 725 University Blvd, Dayton, OH 45435 (scholtz.2@wright.edu).

Author and Disclosure Information

Dr. Scholtz is from the Department of Dermatology, Wright State University, Dayton, Ohio. Dr. Mishra is from the Department of Pathology, Trillium Pathology Inc, Springfield, Ohio. Dr. Simman is from the Wright State University Boonshoft School of Medicine, Dayton, and Jobst Vascular Institute/ProMedica Health System Toledo Hospital, Ohio.

The authors report no conflict of interest.

Correspondence: Jaclyn Scholtz, MD, Department of Dermatology, 725 University Blvd, Dayton, OH 45435 (scholtz.2@wright.edu).

Article PDF
Article PDF

Angiosarcoma is a rare and aggressive vascular malignant neoplasm derived from endothelial cells. In general, sarcomas account for approximately 1% of all malignancies, with approximately 2% being angiosarcomas.1 The risk of recurrence at 5 years is estimated to be 84%, and 5-year survival is estimated at 15% to 30%. Poor prognostic factors for angiosarcoma include large tumor size, depth of invasion greater than 3 mm, high mitotic rate, positive surgical margins, and metastasis.2 Approximately 20% to 40% of patients who are diagnosed with angiosarcoma already have distant metastasis, contributing to the aggressive nature of this neoplasm.3

Angiosarcoma can affect various anatomic locations, including the skin, soft tissue, breasts, and liver. Cutaneous angiosarcoma is the most common clinical manifestation, accounting for approximately 50% to 60% of all cases, and typically is known to occur in 3 distinct settings.2 Primary or idiopathic cutaneous angiosarcoma is most commonly seen in elderly individuals, with a peak incidence in the seventh to eighth decades of life, and presents as a bruiselike lesion predominantly on the head and neck. Angiosarcoma also is seen clinically in patients exposed to radiation treatment, with a median onset of symptoms occurring 5 to 10 years posttreatment, and in patients with chronic lymphedema, usually on the arm following radical mastectomy, which also is known as Stewart-Treves syndrome.2

With any sarcoma, treatment typically first involves surgical excision; however, there is no direct approach for treatment of cutaneous angiosarcoma, as an individual plan typically is needed for each patient. Treatment options include surgical excision, radiation, chemotherapy, or a combination of these therapies.2,4

We present a rare case of cutaneous angiosarcoma of the left leg in the setting of chronic venous insufficiency with some degree of lymphedema and a nonhealing ulcer. This case is unique in that it does not fit the classic presentation of cutaneous angiosarcoma previously described.

Case Report

An 83-year-old woman with a medical history of advanced dementia, congestive heart failure, chronic obstructive pulmonary disease, chronic kidney disease, type 2 diabetes mellitus, hypertension, and chronic venous insufficiency with stasis dermatitis presented to the emergency department following a mechanical fall. Most of her medical history was obtained from the patient’s family. She had a history of multiple falls originally thought to be related to a chronic leg ulcer that had been managed with wound care. Recently, however, the lesion was noted to have increasing erythema surrounding the wound margins. An 8×8-cm erythematous plaque on the anterior lateral left leg with a firm central nodule with hemorrhagic crust that measured approximately 4 cm in diameter was noted by the emergency department physicians (Figure 1). In the emergency department, vitals and other laboratory values were within reference range, and a radiograph of the left tibia/fibula was unremarkable. Cellulitis initially was considered in the emergency department and cephalexin was started; however, since the patient was afebrile and had no leukocytosis, plastic surgery also was consulted. Biopsies were obtained from the superior and inferior parts of the lesion. Histologic analysis revealed a poorly differentiated vascular neoplasm of epithelioid endothelial cells with considerable cell atypia that extended through the entirety of the dermis (Figure 2). The tumor cells stained positive with vimentin and CD34. Pathology noted no immunohistochemistry stains to synaptophysin, S-100, human melanoma black 45, MART-1, CK20, CK7, CK8/18, CK5/6, and p63. The pathologic diagnosis was consistent with cutaneous angiosarcoma. Computed tomography of the chest, abdomen, and pelvis revealed no local or distant metastases.

Figure1
Figure 1. Cutaneous angiosarcoma presenting as a large erythematous plaque on the anterior lateral left lower leg with a firm central nodule with overlying hemorrhagic crust.

Figure2
Figure 2. Histologic analysis revealed a poorly differentiated vascular neoplasm extending through the dermis (A) with epithelioid endothelial cells (B)(H&E, original magnifications ×40 and ×100). Considerable cell atypia and mitotic figures were appreciated on higher power (C)(H&E, original magnification ×400).

A wide excision of the cutaneous angiosarcoma was performed. The initial frozen section analysis revealed positive margins. Three additional excisions still showed positive margins, and further excision was held after obtaining family consent due to the extensive nature of the neoplasm and lengthy operating room time. The final defect after excision measured 15×10×2.5 cm (Figure 3A), and subsequent application of a split-thickness graft was performed. Additional treatment options were discussed with the family, including radiation therapy, amputation of the left lower leg, or no treatment. The family opted not to proceed with further treatment. The graft healed without signs of reoccurrence approximately 3 months later (Figure 3B), and the patient received physical therapy, which allowed her to gain strength and some independence.

Figure3
Figure 3. Wide surgical excision of the cutaneous angiosarcoma yielded a final defect measuring 15×10×2.5 cm (A). Approximately 3 months following excision and subsequent split-thickness skin graft, the patient was healing well with no evidence of reoccurrence (B).

 

 

Comment

Clinical Manifestation
Cutaneous angiosarcoma is a rare malignant vascular neoplasm that when clinically diagnosed is typically seen in 3 settings: (1) idiopathic (commonly on the face and neck), (2) following radiation treatment, and (3) classically following mastectomy with subsequent chronic lymphedema. Our patient did not classically fit these settings of cutaneous angiosarcoma due to the location of the lesion on the lower leg as well as its occurrence in the setting of a chronic nonhealing ulcer and lymphedema.

Chronic lymphedema is a common clinical manifestation that is likely secondary to other medical conditions, such as in our patient. As a result, these patients are at increased risk for developing chronic ulcers due to poor wound healing; however, as seen in our patient, chronic nonhealing ulcers require a broad differential because they may clinically mimic many processes. Patient history and visual presentation were crucial in this case because a biopsy was obtained that ultimately led to the patient’s diagnosis.

Differential Diagnosis
Initially, a venous ulcer secondary to chronic venous insufficiency was considered in the differential for our patient. She had a history of congestive heart failure, kidney disease, and type 2 diabetes mellitus, all of which contribute to lymphedema and/or poor wound healing. However, venous ulcers usually are located on the medial ankles and are irregularly shaped with an erythematous border and fibrinous exudate with central depression, making it a less likely diagnosis in our patient. Additionally, an infectious process was considered, but the patient was afebrile and laboratory values demonstrated no leukocytosis.

Marjolin ulcer was highly suspected because the clinical presentation revealed a nodule with hemorrhagic crust and induration in the setting of a chronic nonhealing ulcer. The pathogenesis of malignancy in chronic ulcers is thought to be due to continuous mitotic activity from regeneration and repair of the wound, especially in the setting of repeated trauma to the area.5 In our patient, the history of multiple falls with possible multitrauma injury to the chronic ulcer further increased suspicion of malignancy. The most common and frequently seen malignancy that develops in chronic ulcers is squamous cell carcinoma (SCC) followed by basal cell carcinoma. Plastic surgery suspected an SCC for the working diagnosis, which prompted a punch biopsy; however, the histologic analysis was not consistent with SCC or basal cell carcinoma. Marjolin ulcer also may demonstrate a periosteal reaction,5 which was not the case with our patient after a radiograph of the left tibia/fibula was unremarkable.

Another potential malignancy to consider is melanoma. There are few case reports of biopsy-proven melanoma from an enlarging chronic ulcer.6,7 Additionally, poorly differentiated angiosarcoma can mimic melanoma2; however, immunohistochemistry stain was negative for S-100, human melanoma black 45, and MART-1, making melanoma unlikely.

Kaposi sarcoma (KS) and angiosarcoma are both malignant vascular tumors that similarly present with red to purple patches, plaques, or nodules, making it difficult to distinguish between the two conditions. It is important to note that KS usually is lower grade, and the pathogenesis is linked to human herpesvirus 8, which can be identified on immunohistochemistry staining. There have been cases of KS reported in patients who have no history of human immunodeficiency virus/AIDS, thus the classic subtype of KS may have been considered in this patient.8 The histologic appearance of KS may vary from dilated irregular endothelial cells lining the vascular space to mild endothelial cell atypia. Histology also shows hemosiderin-laden macrophages, extravasated red blood cells, and an inflammatory infiltrate. An additional malignant vascular neoplasm that needs to be differentiated is epithelioid hemangioendothelioma. Cutaneous presentation of an epithelioid hemangioendothelioma may be similar to what was seen in our patient but histologically will usually show neoplastic cells with pale eosinophilic cytoplasm and vesicular nuclei of plump, oval, polygonal cells in cords or aggregates surrounding vascular channels. These neoplasms also tend to occur around medium- to large-sized veins.1,9 With our patient, even though human herpesvirus 8 was not tested with immunohistochemistry, gold standard immunohistochemistry confirmation with CD34 and vimentin staining combined with poorly differentiated endothelial atypia with mitotic figures on histologic analysis favored angiosarcoma versus KS or epithelioid hemangioendothelioma.10,11

Management
Cutaneous angiosarcoma is a rare and aggressive vascular neoplasm accounting for approximately 2% of all combined sarcomas, with an estimated 20% to 40% having distant metastasis at diagnosis.1,3 For this reason, computed tomography was performed in our patient and revealed no local or distant metastasis. Therefore, chemotherapy was not an appropriate adjuvant treatment option.12 With no evidence of metastasis, initial treatment began with surgical removal but proved to be difficult in our patient. Although the implications of positive surgical margins remain unclear with regard to overall patient survival, surgical resection followed by radiation therapy has been shown to be optimal, as it reduces the risk of local reoccurrence.3 There have been reported cases of cutaneous angiosarcoma of the leg that were treated with amputation without signs of reoccurrence or metastasis.10,13,14 Given the results from these cases and considering that our patient had no metastasis, amputation seemed to be a good prognostic option; however, considering other factors regarding the patient’s comorbidities and quality of life, her family decided not to pursue any further treatment with amputation or radiation therapy.

Conclusion

There should be low threshold for biopsy in patients who present with nonhealing wounds that do not progress in the normal phase of wound healing with suspicion for malignancy. As seen with our patient, cutaneous angiosarcoma can clinically mimic many disease processes, and although rare in nature, it should always be considered when a patient presents with a rapidly growing lesion in the setting of chronic lymphedema or venous ulcer.

Angiosarcoma is a rare and aggressive vascular malignant neoplasm derived from endothelial cells. In general, sarcomas account for approximately 1% of all malignancies, with approximately 2% being angiosarcomas.1 The risk of recurrence at 5 years is estimated to be 84%, and 5-year survival is estimated at 15% to 30%. Poor prognostic factors for angiosarcoma include large tumor size, depth of invasion greater than 3 mm, high mitotic rate, positive surgical margins, and metastasis.2 Approximately 20% to 40% of patients who are diagnosed with angiosarcoma already have distant metastasis, contributing to the aggressive nature of this neoplasm.3

Angiosarcoma can affect various anatomic locations, including the skin, soft tissue, breasts, and liver. Cutaneous angiosarcoma is the most common clinical manifestation, accounting for approximately 50% to 60% of all cases, and typically is known to occur in 3 distinct settings.2 Primary or idiopathic cutaneous angiosarcoma is most commonly seen in elderly individuals, with a peak incidence in the seventh to eighth decades of life, and presents as a bruiselike lesion predominantly on the head and neck. Angiosarcoma also is seen clinically in patients exposed to radiation treatment, with a median onset of symptoms occurring 5 to 10 years posttreatment, and in patients with chronic lymphedema, usually on the arm following radical mastectomy, which also is known as Stewart-Treves syndrome.2

With any sarcoma, treatment typically first involves surgical excision; however, there is no direct approach for treatment of cutaneous angiosarcoma, as an individual plan typically is needed for each patient. Treatment options include surgical excision, radiation, chemotherapy, or a combination of these therapies.2,4

We present a rare case of cutaneous angiosarcoma of the left leg in the setting of chronic venous insufficiency with some degree of lymphedema and a nonhealing ulcer. This case is unique in that it does not fit the classic presentation of cutaneous angiosarcoma previously described.

Case Report

An 83-year-old woman with a medical history of advanced dementia, congestive heart failure, chronic obstructive pulmonary disease, chronic kidney disease, type 2 diabetes mellitus, hypertension, and chronic venous insufficiency with stasis dermatitis presented to the emergency department following a mechanical fall. Most of her medical history was obtained from the patient’s family. She had a history of multiple falls originally thought to be related to a chronic leg ulcer that had been managed with wound care. Recently, however, the lesion was noted to have increasing erythema surrounding the wound margins. An 8×8-cm erythematous plaque on the anterior lateral left leg with a firm central nodule with hemorrhagic crust that measured approximately 4 cm in diameter was noted by the emergency department physicians (Figure 1). In the emergency department, vitals and other laboratory values were within reference range, and a radiograph of the left tibia/fibula was unremarkable. Cellulitis initially was considered in the emergency department and cephalexin was started; however, since the patient was afebrile and had no leukocytosis, plastic surgery also was consulted. Biopsies were obtained from the superior and inferior parts of the lesion. Histologic analysis revealed a poorly differentiated vascular neoplasm of epithelioid endothelial cells with considerable cell atypia that extended through the entirety of the dermis (Figure 2). The tumor cells stained positive with vimentin and CD34. Pathology noted no immunohistochemistry stains to synaptophysin, S-100, human melanoma black 45, MART-1, CK20, CK7, CK8/18, CK5/6, and p63. The pathologic diagnosis was consistent with cutaneous angiosarcoma. Computed tomography of the chest, abdomen, and pelvis revealed no local or distant metastases.

Figure1
Figure 1. Cutaneous angiosarcoma presenting as a large erythematous plaque on the anterior lateral left lower leg with a firm central nodule with overlying hemorrhagic crust.

Figure2
Figure 2. Histologic analysis revealed a poorly differentiated vascular neoplasm extending through the dermis (A) with epithelioid endothelial cells (B)(H&E, original magnifications ×40 and ×100). Considerable cell atypia and mitotic figures were appreciated on higher power (C)(H&E, original magnification ×400).

A wide excision of the cutaneous angiosarcoma was performed. The initial frozen section analysis revealed positive margins. Three additional excisions still showed positive margins, and further excision was held after obtaining family consent due to the extensive nature of the neoplasm and lengthy operating room time. The final defect after excision measured 15×10×2.5 cm (Figure 3A), and subsequent application of a split-thickness graft was performed. Additional treatment options were discussed with the family, including radiation therapy, amputation of the left lower leg, or no treatment. The family opted not to proceed with further treatment. The graft healed without signs of reoccurrence approximately 3 months later (Figure 3B), and the patient received physical therapy, which allowed her to gain strength and some independence.

Figure3
Figure 3. Wide surgical excision of the cutaneous angiosarcoma yielded a final defect measuring 15×10×2.5 cm (A). Approximately 3 months following excision and subsequent split-thickness skin graft, the patient was healing well with no evidence of reoccurrence (B).

 

 

Comment

Clinical Manifestation
Cutaneous angiosarcoma is a rare malignant vascular neoplasm that when clinically diagnosed is typically seen in 3 settings: (1) idiopathic (commonly on the face and neck), (2) following radiation treatment, and (3) classically following mastectomy with subsequent chronic lymphedema. Our patient did not classically fit these settings of cutaneous angiosarcoma due to the location of the lesion on the lower leg as well as its occurrence in the setting of a chronic nonhealing ulcer and lymphedema.

Chronic lymphedema is a common clinical manifestation that is likely secondary to other medical conditions, such as in our patient. As a result, these patients are at increased risk for developing chronic ulcers due to poor wound healing; however, as seen in our patient, chronic nonhealing ulcers require a broad differential because they may clinically mimic many processes. Patient history and visual presentation were crucial in this case because a biopsy was obtained that ultimately led to the patient’s diagnosis.

Differential Diagnosis
Initially, a venous ulcer secondary to chronic venous insufficiency was considered in the differential for our patient. She had a history of congestive heart failure, kidney disease, and type 2 diabetes mellitus, all of which contribute to lymphedema and/or poor wound healing. However, venous ulcers usually are located on the medial ankles and are irregularly shaped with an erythematous border and fibrinous exudate with central depression, making it a less likely diagnosis in our patient. Additionally, an infectious process was considered, but the patient was afebrile and laboratory values demonstrated no leukocytosis.

Marjolin ulcer was highly suspected because the clinical presentation revealed a nodule with hemorrhagic crust and induration in the setting of a chronic nonhealing ulcer. The pathogenesis of malignancy in chronic ulcers is thought to be due to continuous mitotic activity from regeneration and repair of the wound, especially in the setting of repeated trauma to the area.5 In our patient, the history of multiple falls with possible multitrauma injury to the chronic ulcer further increased suspicion of malignancy. The most common and frequently seen malignancy that develops in chronic ulcers is squamous cell carcinoma (SCC) followed by basal cell carcinoma. Plastic surgery suspected an SCC for the working diagnosis, which prompted a punch biopsy; however, the histologic analysis was not consistent with SCC or basal cell carcinoma. Marjolin ulcer also may demonstrate a periosteal reaction,5 which was not the case with our patient after a radiograph of the left tibia/fibula was unremarkable.

Another potential malignancy to consider is melanoma. There are few case reports of biopsy-proven melanoma from an enlarging chronic ulcer.6,7 Additionally, poorly differentiated angiosarcoma can mimic melanoma2; however, immunohistochemistry stain was negative for S-100, human melanoma black 45, and MART-1, making melanoma unlikely.

Kaposi sarcoma (KS) and angiosarcoma are both malignant vascular tumors that similarly present with red to purple patches, plaques, or nodules, making it difficult to distinguish between the two conditions. It is important to note that KS usually is lower grade, and the pathogenesis is linked to human herpesvirus 8, which can be identified on immunohistochemistry staining. There have been cases of KS reported in patients who have no history of human immunodeficiency virus/AIDS, thus the classic subtype of KS may have been considered in this patient.8 The histologic appearance of KS may vary from dilated irregular endothelial cells lining the vascular space to mild endothelial cell atypia. Histology also shows hemosiderin-laden macrophages, extravasated red blood cells, and an inflammatory infiltrate. An additional malignant vascular neoplasm that needs to be differentiated is epithelioid hemangioendothelioma. Cutaneous presentation of an epithelioid hemangioendothelioma may be similar to what was seen in our patient but histologically will usually show neoplastic cells with pale eosinophilic cytoplasm and vesicular nuclei of plump, oval, polygonal cells in cords or aggregates surrounding vascular channels. These neoplasms also tend to occur around medium- to large-sized veins.1,9 With our patient, even though human herpesvirus 8 was not tested with immunohistochemistry, gold standard immunohistochemistry confirmation with CD34 and vimentin staining combined with poorly differentiated endothelial atypia with mitotic figures on histologic analysis favored angiosarcoma versus KS or epithelioid hemangioendothelioma.10,11

Management
Cutaneous angiosarcoma is a rare and aggressive vascular neoplasm accounting for approximately 2% of all combined sarcomas, with an estimated 20% to 40% having distant metastasis at diagnosis.1,3 For this reason, computed tomography was performed in our patient and revealed no local or distant metastasis. Therefore, chemotherapy was not an appropriate adjuvant treatment option.12 With no evidence of metastasis, initial treatment began with surgical removal but proved to be difficult in our patient. Although the implications of positive surgical margins remain unclear with regard to overall patient survival, surgical resection followed by radiation therapy has been shown to be optimal, as it reduces the risk of local reoccurrence.3 There have been reported cases of cutaneous angiosarcoma of the leg that were treated with amputation without signs of reoccurrence or metastasis.10,13,14 Given the results from these cases and considering that our patient had no metastasis, amputation seemed to be a good prognostic option; however, considering other factors regarding the patient’s comorbidities and quality of life, her family decided not to pursue any further treatment with amputation or radiation therapy.

Conclusion

There should be low threshold for biopsy in patients who present with nonhealing wounds that do not progress in the normal phase of wound healing with suspicion for malignancy. As seen with our patient, cutaneous angiosarcoma can clinically mimic many disease processes, and although rare in nature, it should always be considered when a patient presents with a rapidly growing lesion in the setting of chronic lymphedema or venous ulcer.

References
  1. Kumar V, Abbas A, Aster J. Robbins Basic Pathology. 9th ed. Philadelphia, PA: Elsevier Saunders; 2013.
  2. Donghi D, Kerl K, Dummer R, et al. Cutaneous angiosarcoma: own experience over 13 years. clinical features, disease course and immunohistochemical profile. J Eur Acad Dermatol Venereol. 2010;24:1230-1234.
  3. Dossett LA, Harrington M, Cruse CW, et al. Cutaneous angiosarcoma. Curr Probl Cancer. 2015;39:258-263.
  4. Morgan MB, Swann M, Somach S, et al. Cutaneous angiosarcoma: a case series with prognostic correlation. J Am Acad Dermatol. 2004;50:867-874.
  5. Pekarek B, Buck S, Osher L. A comprehensive review on Marjolin’s ulcers: diagnosis and treatment. J Am Col Certif Wound Spec. 2011;3:60-64.
  6. Gerslova A, Pokorna A, Stukavcova A, et al. Rare cause of non-healing foot wound—acral lentiginous melanoma. Neuro Endocrinol Lett. 2012;37:12-17.
  7. Turk BG, Bozkurt A, Yaman B, et al. Melanoma arising in chronic ulceration associated with lymphoedema. J Wound Care. 2013;22:74-75.
  8. Phavixay L, Raynolds D, Simman R. Non AIDS Kaposi’s sarcoma leading to lower extremities wounds, case presentations and discussion.J Am Coll Clin Wound Spec. 2012;4:13-15.
  9. Requena L, Kutzner H. Hemangioendothelioma. Semin Diagn Pathol. 2013;30:29-44.
  10. Harrison WD, Chandrasekar CR. Stewart-Treves syndrome following idiopathic leg lymphoedema: remember sarcoma. J Wound Care. 2015;24(6 suppl):S5-S7.
  11. Kak I, Salama S, Gohla G, et al. A case of patch stage of Kaposi’s sarcoma and discussion of the differential diagnosis. Rare Tumors. 2016;8:6123.
  12. Agulnik M, Yarber JL, Okuno SH, et al. An open-label, multicenter, phase II study of bevacizumab for the treatment of angiosarcoma and epithelioid hemangioendotheliomas. Ann Oncol. 2013;24:257-263.
  13. Linda DD, Harish S, Alowami S, et al. Radiology-pathology conference: cutaneous angiosarcoma of the leg. Clin Imaging. 2013;37:602-607.
  14. Roy P, Clark MA, Thomas JM. Stewart-Treves syndrome—treatment and outcome in six patients from a single centre. Eur J Surg Oncol. 2004;30:982-986.
References
  1. Kumar V, Abbas A, Aster J. Robbins Basic Pathology. 9th ed. Philadelphia, PA: Elsevier Saunders; 2013.
  2. Donghi D, Kerl K, Dummer R, et al. Cutaneous angiosarcoma: own experience over 13 years. clinical features, disease course and immunohistochemical profile. J Eur Acad Dermatol Venereol. 2010;24:1230-1234.
  3. Dossett LA, Harrington M, Cruse CW, et al. Cutaneous angiosarcoma. Curr Probl Cancer. 2015;39:258-263.
  4. Morgan MB, Swann M, Somach S, et al. Cutaneous angiosarcoma: a case series with prognostic correlation. J Am Acad Dermatol. 2004;50:867-874.
  5. Pekarek B, Buck S, Osher L. A comprehensive review on Marjolin’s ulcers: diagnosis and treatment. J Am Col Certif Wound Spec. 2011;3:60-64.
  6. Gerslova A, Pokorna A, Stukavcova A, et al. Rare cause of non-healing foot wound—acral lentiginous melanoma. Neuro Endocrinol Lett. 2012;37:12-17.
  7. Turk BG, Bozkurt A, Yaman B, et al. Melanoma arising in chronic ulceration associated with lymphoedema. J Wound Care. 2013;22:74-75.
  8. Phavixay L, Raynolds D, Simman R. Non AIDS Kaposi’s sarcoma leading to lower extremities wounds, case presentations and discussion.J Am Coll Clin Wound Spec. 2012;4:13-15.
  9. Requena L, Kutzner H. Hemangioendothelioma. Semin Diagn Pathol. 2013;30:29-44.
  10. Harrison WD, Chandrasekar CR. Stewart-Treves syndrome following idiopathic leg lymphoedema: remember sarcoma. J Wound Care. 2015;24(6 suppl):S5-S7.
  11. Kak I, Salama S, Gohla G, et al. A case of patch stage of Kaposi’s sarcoma and discussion of the differential diagnosis. Rare Tumors. 2016;8:6123.
  12. Agulnik M, Yarber JL, Okuno SH, et al. An open-label, multicenter, phase II study of bevacizumab for the treatment of angiosarcoma and epithelioid hemangioendotheliomas. Ann Oncol. 2013;24:257-263.
  13. Linda DD, Harish S, Alowami S, et al. Radiology-pathology conference: cutaneous angiosarcoma of the leg. Clin Imaging. 2013;37:602-607.
  14. Roy P, Clark MA, Thomas JM. Stewart-Treves syndrome—treatment and outcome in six patients from a single centre. Eur J Surg Oncol. 2004;30:982-986.
Issue
Cutis - 102(4)
Issue
Cutis - 102(4)
Page Number
E8-E11
Page Number
E8-E11
Publications
Publications
Topics
Article Type
Display Headline
Cutaneous Angiosarcoma of the Lower Leg
Display Headline
Cutaneous Angiosarcoma of the Lower Leg
Sections
Inside the Article

Practice Points

  • Cutaneous angiosarcoma is a rare malignant vascular neoplasm typically seen in 3 settings: (1) idiopathic (commonly on the face and neck), (2) following radiation treatment, and (3) classically in the setting of chronic lymphedema following mastectomy (Stewart-Treves syndrome).
  • There should be a low threshold for biopsy in patients who present with nonhealing wounds that do not progress in the normal phase of wound healing with suspicion for malignancy.
  • Histologic analysis of angiosarcoma shows positive staining for CD34 and vimentin with poorly differentiated endothelial atypia with mitotic figures.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Article PDF Media

Primary Cutaneous Apocrine Carcinoma Arising Within a Nevus Sebaceus

Article Type
Changed
Thu, 01/10/2019 - 13:54

Nevus sebaceus (NS) is a benign hair follicle neoplasm present in approximately 1.3% of the population, typically involving the scalp, neck, or face.1 These lesions usually are present at birth or identified soon after, during the first year. They present as a yellowish hairless patch or plaque but can develop a more papillomatous appearance, especially after puberty. Historically, the concern with NS was its tendency to transform into basal cell carcinoma (BCC), which prompted surgical excision of the lesion during childhood. This theory has been discounted more recently, as further research has suggested that what was once thought to be BCC may have been confused with the similarly appearing trichoblastoma; however, malignant transformation of NS does still occur, with BCC still being the most common.2 We present the case of a long-standing NS with rare transformation to apocrine carcinoma.

Case Report

A 76-year-old woman presented with several new lesions within a previously diagnosed NS. She reported having the large plaque for as long as she could recall but reported that several new growths developed within the plaque over the last 2 months, slowly increasing in size. She reported a prior biopsy within the growth several years prior, which she described as an irritated seborrheic keratosis.

Physical examination demonstrated 4 distinct lesions within the flesh-colored, verrucous plaque located on the left side of the temporal scalp (Figure 1). The first lesion was a 2.5-cm pearly, pink, exophytic tumor (labeled as A in Figure 1). The next 2 lesions were brown, pedunculated, verrucous papules (labeled as B and C in Figure 1). The last lesion was a purple papule (labeled as D in Figure 1). Four shave biopsies were performed for histologic analysis of the lesions. Lesions B, C, and D were consistent with trichoblastomas, as pathology showed basaloid epithelial tumors that displayed primitive follicular structures, areas of stromal induction, and some pigmentation. Lesion A, originally thought to be suspicious for a BCC, was determined to be a primary cutaneous apocrine adenocarcinoma upon pathologic review. The pathology showed a dermal tumor displaying solid and tubular areas with decapitation secretion. Nuclear pleomorphism and mitoses were present (Figure 2), and staining for carcinoembryonic antigen was positive (Figure 3). Immunoreactivity with epithelial membrane antigen and cytokeratin 7 was noted as well as focal positivity for mammaglobin. Primary apocrine carcinoma was favored over metastatic carcinoma due to the location of the lesion within an NS along with a negative history of internal malignancy. Dermatopathology recommended complete removal of all lesions within the NS.

Figure1
Figure 1. Verrucous plaque and 4 distinct neoplasms (labeled A–D) on the left side of the temporal scalp.

Figure2
Figure 2. Apocrine carcinoma histopathology demonstrating solid and tubular areas with decapitation secretion (A)(H&E). On high power, marked nuclear pleomorphism and increased mitotic activity were observed (B)(H&E).

Figure3
Figure 3. Apocrine carcinoma demonstrating carcinoembryonic antigen staining positivity.


Upon discussing biopsy results and recommendations with our patient, she agreed to undergo excision with intraoperative pathology by a plastic surgeon within our practice to ensure clear margins. The surgical defect following excision was sizeable and closed utilizing a rhomboid flap, full-thickness skin graft, and a split-thickness skin graft. At surgical follow-up, she was doing well and there have been no signs of local recurrence for 10 months since excision.

 

 

Comment

Presentation
Nevus sebaceus is the most common adnexal tumor and is classified as a benign congenital hair follicle tumor that is located most commonly on the scalp but also occurs on the face and neck.1 The lesions usually are present at birth but also can develop during the first year of life.2 Diagnosis may be later, during adolescence, when patients seek medical attention during the lesion’s rapid growth phase.1 Nevus sebaceus also is known as an organoid nevus because it may contain all components of the skin. It was originally identified by Jadassohn in 1895.3 It presents as a yellowish, smooth, hairless patch or plaque in prepubertal patients. During adolescence, the lesion typically becomes more yellowish, as well as papillomatous, scaly, or warty. The reported incidence of NS is 0.05% to 1% in dermatology patients.2

Differential
Nevus sebaceus also is a component of several syndromes that should be kept in mind, including Schimmelpenning-Feuerstein-Mims syndrome, which presents with neurologic, skeletal, genitourinary, cardiovascular, and ophthalmic disorders, in addition to cutaneous features. Others include phacomatosis pigmentokeratotica, didmyosis aplasticosebacea, SCALP syndrome (sebaceus nevus, central nervous system malformations, aplasia cutis congenita, limbal dermoid, and pigmented nevus), and more.4,5

Etiology
The etiology of NS has not been completely determined. One study that evaluated 44 NS tissue samples suggested the presence of human papillomavirus (HPV) in NS formation, finding that 82% of NS lesions studied contained HPV DNA. From these results, Carlson et al6 suggested a possible maternal transmission of HPV and infection of ectodermal cells as a potential cause of NS; however, this hypothesis was soon challenged by a study that showed a complete absence of HPV in 16 samples via histological evaluation and polymerase chain reaction for a broad range of HPV types.7 There were investigations into a patched (PTCH) deletion as the cause of NS and thus explained the historically high rate of secondary BCC.8 Further studies showed no mutations at the PTCH locus in trichoblastomas or other tumors arising from NS.9,10

More recent studies have recognized HRAS and KRAS mutations as a causative factor in NS.11 Nevus sebaceus belongs to a group of syndromes resulting from lethal mutations that survive via mosaicism. Nevus sebaceus is caused by postzygotic HRAS or KRAS mutations and is known as a mosaic RASopathy.12 In fact, there is growing evidence to suggest that other nevoid proliferations including keratinocytic epidermal nevi and melanocytic nevi also fall into the spectrum of mosaic RASopathies.13

Staging
There are 3 clinical stages of NS, originally described by Mehregan and Pinkus.14 In stage I (historically known as the infantile stage), the lesion presents as a yellow to pink, smooth, hairless patch. Histologic features include immature hair follicles and hypoplastic sebaceous glands. In stage II (also known as the puberty stage), the lesion becomes more pronounced. Firmer plaques can develop with hyperkeratosis. Hormonal changes cause sebaceous glands to develop, accompanied by epidermal hyperplasia and maturation of apocrine glands. Stage III (the tumoral stage) is a period that various neoplasms have the highest likelihood of occurring. Nevus sebaceus in an adolescent or adult demonstrates mature adnexal structures and greater epidermal hyperplasia.2,4,15

Malignancy
By virtue of these stages of NS development, malignant transformation is expected most often during stage III. However, cases have been reported of malignant tumor development in NS in children before puberty. Two case reports described a 7-year-old boy and a 10-year-old boy diagnosed with a BCC arising from an NS.16,17 However, secondary BCC formation before 16 years of age is rare. Basal cell carcinoma arising from an NS has been commonly reported and is the most common malignant neoplasm in NS (1.1%).2,3 However, the most common neoplasm overall is trichoblastoma (7.4%). The second most common tumor was syringocystadenoma papilliferum, occurring in approximately 5.2% of NS cases. The neoplasm rate in NS was found to be proportional to the patient age.2,18 Multiple studies have shown the overall rate of secondary neoplasms in NS to be 13% to 21.4%, with malignant tumors composing 0.8% to 2.5%.2,15,19 Other neoplasms that have been reported include keratoacanthoma, trichilemmoma, sebaceoma, nevocellular nevus, squamous cell carcinoma, adnexal carcinoma, apocrine adenocarcinoma, and malignant melanoma.19-21

It is argued that the reported rate of BCC formation is overestimated, as prior studies incorrectly labeled trichoblastomas as BCCs. In fact, the largest studies of NS from the 1990s revealed lower rates of malignant secondary tumors than previously determined.4

The identification of apocrine adenocarcinoma tumors arising from NS is exceedingly rare. A study performed by Cribier et al19 in 2000 retrospectively analyzed 596 cases of excised NS from 1932 to 1998. No apocrine carcinomas were reported in this study.19 Approximately 12 cases have been previously reported throughout the literature.20-26 Apocrine carcinomas occur most frequently in apocrine-rich areas such as the axillae, external ears, eyelids, and anogenital area. However, in the cases with apocrine carcinomas that developed from NS, the carcinomas have been located almost exclusively on the scalp.23

Histopathology
Histopathologic examination reveals considerable variation in morphology, and an underlying pattern has been difficult to recognize. Unfortunately, some authors have concluded that the diagnosis of apocrine carcinoma is relatively subjective.26 Robson et al26 identified 3 general architectural patterns: tubular, tubulopapillary, and solid. Tubular structures consisted of glands and ducts lined by a single or multilayered epithelium. Tubulopapillary architecture was characterized by epithelium forming papillary folds without a fibrovascular core. The solid morphology showed sheets of cells with limited ductal or tubular formation.26 The most specific criteria of these apocrine carcinomas are identification of decapitation secretion, periodic acid–Schiff–positive diastase-resistant material present in the cells or lumen, and positive immunostaining for gross cystic disease fluid protein-15.27

Robson et al26 reported estrogen receptor positivity and androgen receptor positivity in 62% and 64% of 24 primary apocrine carcinoma cases, respectively. However, whether these markers are as common in NS-related apocrine carcinomas has yet to be noted in the literature. One study reports a case of apocrine carcinoma from NS with positive staining for human epidermal growth factor-2, a cell membrane receptor tyrosine kinase commonly investigated in breast cancers and extramammary Paget disease.22

These apocrine carcinomas do have the potential for lymphatic metastasis, as seen with multiple studies. Domingo and Helwig21 identified regional lymph node metastasis in 2 of its 4 apocrine carcinoma patients. Robson et al26 reported lymphovascular invasion in 4 cases and perineural invasion in 2 of 24 patients studied. However, even in the context of recurrence and regional metastasis, the prognosis was good and seldom fatal.26

Treatment
The most effective treatment of NS is excision of dermal and epidermal components. Excision should be completed with a minimum of 2- to 3-mm margins and full thickness down to the underlying supporting fat.28 Historically, the practice of prophylactic excision of NS was supported by the potential for malignant transformation; however, early excision of NS may be less reasonable in light of these more recent studies showing lower incidence of BCC (0.8%), replaced by benign trichoblastomas.19 In the case of apocrine carcinoma development, excision is undoubtedly recommended, with unclear recommendations regarding further evaluation for metastasis.

Excision also may be favored for cosmetic purposes, given the visible regions where NS tends to develop. Chepla and Gosain29 argued that surgical intervention should be based on other factors such as location on the scalp, alopecia, and other issues affecting appearance and monitoring rather than incidence of malignant transformation. Close monitoring and biopsy of suspicious areas is a more conservative option.

Other therapies include CO2 laser, as demonstrated by Kiedrowicz et al,30 on linear NS in a patient with Schimmelpenning-Feuerstein-Mims syndrome.31 However, this approach is palliative and not effective in removing the entire lesion. Electrodesiccation and curettage and dermabrasion also are not good options for the same reason.4

Occurrence in Children
Nevus sebaceus in children, accompanied by other findings suggestive of epidermal nevus syndromes, should prompt further investigation. Schimmelpenning-Feuerstein-Mims syndrome includes major neurological abnormalities including hemimegalencephaly and seizures.32

Conclusion

Apocrine carcinomas are malignant neoplasms that may rarely arise within an NS. Their clinical identification is difficult and requires histopathologic evaluation. Upon recognition, prompt excision with tumor-free margins is recommended. As a rare entity, little data is available regarding its metastatic potential or overall survival rates. Further investigation is clearly necessary as new cases arise.

References
  1. Kamyab-Hesari K, Balochi K, Afshar N, et al. Clinicopathological study of 1016 consecutive adnexal skin tumors. Acta Med Iran. 2013;51:879-885.
  2. Idriss MH, Elston DM. Secondary neoplasms associated with nevus sebaceus of Jadassohn: a study of 707 cases. J Am Acad Dermatol. 2014;70:332-337.
  3. Ball EA, Hussain M, Moss AL. Squamous cell carcinoma and basal cell carcinoma arising in a naevus sebaceous of Jadassohn: case report and literature review. Clin Exp Dermatol. 2005;30:259-260.
  4. Moody MN, Landau JM, Goldberg LH. Nevus sebaceous revisited. Pediatr Dermatol. 2012;29:15-23.
  5. Happle R. The group of epidermal nevus syndromes part I. well defined phenotypes. J Am Acad Dermatol. 2010;63:1-22; quiz 23-24.
  6. Carlson JA, Cribier B, Nuovo G, et al. Epidermodysplasia verruciformis-associated and genital-mucosal high-risk human papillomavirus DNA are prevalent in nevus sebaceus of Jadassohn. J Am Acad Dermatol. 2008;59:279-294.
  7. Kim D, Benjamin LT, Sahoo MK, et al. Human papilloma virus is not prevalent in nevus sebaceus [published online November 14, 2013]. Pediatr Dermatol. 2014;31:326-330.
  8. Xin H, Matt D, Qin JZ, et al. The sebaceous nevus: a nevus with deletions of the PTCH gene. Cancer Res. 1999;59:1834-1836.
  9. Hafner C, Schmiemann V, Ruetten A, et al. PTCH mutations are not mainly involved in the pathogenesis of sporadic trichoblastomas. Hum Pathol. 2007;38:1496-1500.
  10. Takata M, Tojo M, Hatta N, et al. No evidence of deregulated patched-hedgehog signaling pathway in trichoblastomas and other tumors arising within nevus sebaceous. J Invest Dermatol. 2001;117:1666-1670.
  11. Levinsohn JL, Tian LC, Boyden LM, et al. Whole-exome sequencing reveals somatic mutations in HRAS and KRAS, which cause nevus sebaceus [published online October 25, 2012]. J Invest Dermatol. 2013;133:827-830.
  12. Happle R. Nevus sebaceus is a mosaic RASopathy. J Invest Dermatol. 2013;133:597-600.
  13. Luo S, Tsao H. Epidermal, sebaceous, and melanocytic nevoid proliferations are spectrums of mosaic RASopathies. J Invest Dermatol. 2014;134:2493-2496.
  14. Mehregan AH, Pinkus H. Life history of organoid nevi. special reference to nevus sebaceus of Jadassohn. Arch Dermatol. 1965;91:574-588.
  15. Muñoz-Pérez MA, García-Hernandez MJ, Ríos JJ, et al. Sebaceus naevi: a clinicopathologic study. J Eur Acad Dermatol Venereol. 2002;16:319-324.
  16. Altaykan A, Ersoy-Evans S, Erkin G, et al. Basal cell carcinoma arising in nevus sebaceous during childhood. Pediatr Dermatol. 2008;25:616-619.
  17. Turner CD, Shea CR, Rosoff PM. Basal cell carcinoma originating from a nevus sebaceus on the scalp of a 7-year-old boy. J Pediatr Hematol Oncol. 2001;23:247-249.
  18. Jaqueti G, Requena L, Sánchez Yus E. Trichoblastoma is the most common neoplasm developed in nevus sebaceus of Jadassohn: a clinicopathologic study of a series of 155 cases. Am J Dermatopathol. 2000;22:108-118.
  19. Cribier B, Scrivener Y, Grosshans E. Tumors arising in nevus sebaceus: a study of 596 cases. J Am Acad Dermatol. 2000;42(2, pt 1):263-268.
  20. Paudel U, Jha A, Pokhrel DB, et al. Apocrine carcinoma developing in a naevus sebaceous of scalp. Kathmandu Univ Med J (KUMJ). 2012;10:103-105.
  21. Domingo J, Helwig EB. Malignant neoplasms associated with nevus sebaceus of Jadassohn. J Am Acad Dermatol. 1979;1:545-556.
  22. Tanese K, Wakabayashi A, Suzuki T, et al. Immunoexpression of human epidermal growth factor receptor-2 in apocrine carcinoma arising in naevus sebaceous, case report [published online August 23, 2009]. J Eur Acad Dermatol Venereol. 2010;24:360-362.
  23. Dalle S, Skowron F, Balme B, et al. Apocrine carcinoma developed in nevus sebaceus of Jadassohn. Eur J Dermatol. 2003;13:487-489.
  24. Jacyk WK, Requena L, Sánchez Yus E, et al. Tubular apocrine carcinoma arising in a nevus sebaceus of Jadassohn. Am J Dermatopathol. 1998;20:389-392.
  25. Ansai S, Koseki S, Hashimoto H, et al. A case of ductal sweat gland carcinoma connected to syringocystadenoma papilliferum arising in nevus sebaceus. J Cutan Pathol. 1994;21:557-563.
  26. Robson A, Lazar AJ, Ben Nagi J, et al. Primary cutaneous apocrine carcinoma: a clinico-pathologic analysis of 24 cases. Am J Surg Pathol. 2008;32:682-690.
  27. Paties C, Taccagni GL, Papotti M, et al. Apocrine carcinoma of the skin. a clinicopathologic, immunocytochemical, and ultrastructural study. Cancer. 1993;71:375-381.
  28. Davison SP, Khachemoune A, Yu D, et al. Nevus sebaceus of Jadassohn revisited with reconstruction options. Int J Dermatol. 2005;44:145-150.
  29. Chepla KJ, Gosain AK. Giant nevus sebaceus: definition, surgical techniques, and rationale for treatment. Plast Reconstr Surg. 2012;130:296E-304E.
  30. Kiedrowicz M, Kacalak-Rzepka A, Królicki A et al. Therapeutic effects of CO2 laser therapy of linear nevus sebaceous in the course of the Schimmelpenning-Feuerstein-Mims syndrome. Postepy Dermatol Allergol. 2013;30:320-323.
  31. Ashinoff R. Linear nevus sebaceus of Jadassohn treated with the carbon dioxide laser. Pediatr Dermatol. 1993;10:189-191.
  32. van de Warrenburg BP, van Gulik S, Renier WO, et al. The linear naevus sebaceus syndrome. Clin Neurol Neurosurg. 1998;100:126-132.
Article PDF
Author and Disclosure Information

From Nova Southeastern University College of Osteopathic Medicine/Largo Medical Center, Florida.

The authors report no conflict of interest.

Correspondence: Natalie Edgar, DO, Largo Medical Center, Graduate Medical Education, 201 14th St SW, Largo, FL 33770 (natalie.edgar@okstate.edu).

Issue
Cutis - 102(4)
Publications
Topics
Page Number
291-294
Sections
Author and Disclosure Information

From Nova Southeastern University College of Osteopathic Medicine/Largo Medical Center, Florida.

The authors report no conflict of interest.

Correspondence: Natalie Edgar, DO, Largo Medical Center, Graduate Medical Education, 201 14th St SW, Largo, FL 33770 (natalie.edgar@okstate.edu).

Author and Disclosure Information

From Nova Southeastern University College of Osteopathic Medicine/Largo Medical Center, Florida.

The authors report no conflict of interest.

Correspondence: Natalie Edgar, DO, Largo Medical Center, Graduate Medical Education, 201 14th St SW, Largo, FL 33770 (natalie.edgar@okstate.edu).

Article PDF
Article PDF

Nevus sebaceus (NS) is a benign hair follicle neoplasm present in approximately 1.3% of the population, typically involving the scalp, neck, or face.1 These lesions usually are present at birth or identified soon after, during the first year. They present as a yellowish hairless patch or plaque but can develop a more papillomatous appearance, especially after puberty. Historically, the concern with NS was its tendency to transform into basal cell carcinoma (BCC), which prompted surgical excision of the lesion during childhood. This theory has been discounted more recently, as further research has suggested that what was once thought to be BCC may have been confused with the similarly appearing trichoblastoma; however, malignant transformation of NS does still occur, with BCC still being the most common.2 We present the case of a long-standing NS with rare transformation to apocrine carcinoma.

Case Report

A 76-year-old woman presented with several new lesions within a previously diagnosed NS. She reported having the large plaque for as long as she could recall but reported that several new growths developed within the plaque over the last 2 months, slowly increasing in size. She reported a prior biopsy within the growth several years prior, which she described as an irritated seborrheic keratosis.

Physical examination demonstrated 4 distinct lesions within the flesh-colored, verrucous plaque located on the left side of the temporal scalp (Figure 1). The first lesion was a 2.5-cm pearly, pink, exophytic tumor (labeled as A in Figure 1). The next 2 lesions were brown, pedunculated, verrucous papules (labeled as B and C in Figure 1). The last lesion was a purple papule (labeled as D in Figure 1). Four shave biopsies were performed for histologic analysis of the lesions. Lesions B, C, and D were consistent with trichoblastomas, as pathology showed basaloid epithelial tumors that displayed primitive follicular structures, areas of stromal induction, and some pigmentation. Lesion A, originally thought to be suspicious for a BCC, was determined to be a primary cutaneous apocrine adenocarcinoma upon pathologic review. The pathology showed a dermal tumor displaying solid and tubular areas with decapitation secretion. Nuclear pleomorphism and mitoses were present (Figure 2), and staining for carcinoembryonic antigen was positive (Figure 3). Immunoreactivity with epithelial membrane antigen and cytokeratin 7 was noted as well as focal positivity for mammaglobin. Primary apocrine carcinoma was favored over metastatic carcinoma due to the location of the lesion within an NS along with a negative history of internal malignancy. Dermatopathology recommended complete removal of all lesions within the NS.

Figure1
Figure 1. Verrucous plaque and 4 distinct neoplasms (labeled A–D) on the left side of the temporal scalp.

Figure2
Figure 2. Apocrine carcinoma histopathology demonstrating solid and tubular areas with decapitation secretion (A)(H&E). On high power, marked nuclear pleomorphism and increased mitotic activity were observed (B)(H&E).

Figure3
Figure 3. Apocrine carcinoma demonstrating carcinoembryonic antigen staining positivity.


Upon discussing biopsy results and recommendations with our patient, she agreed to undergo excision with intraoperative pathology by a plastic surgeon within our practice to ensure clear margins. The surgical defect following excision was sizeable and closed utilizing a rhomboid flap, full-thickness skin graft, and a split-thickness skin graft. At surgical follow-up, she was doing well and there have been no signs of local recurrence for 10 months since excision.

 

 

Comment

Presentation
Nevus sebaceus is the most common adnexal tumor and is classified as a benign congenital hair follicle tumor that is located most commonly on the scalp but also occurs on the face and neck.1 The lesions usually are present at birth but also can develop during the first year of life.2 Diagnosis may be later, during adolescence, when patients seek medical attention during the lesion’s rapid growth phase.1 Nevus sebaceus also is known as an organoid nevus because it may contain all components of the skin. It was originally identified by Jadassohn in 1895.3 It presents as a yellowish, smooth, hairless patch or plaque in prepubertal patients. During adolescence, the lesion typically becomes more yellowish, as well as papillomatous, scaly, or warty. The reported incidence of NS is 0.05% to 1% in dermatology patients.2

Differential
Nevus sebaceus also is a component of several syndromes that should be kept in mind, including Schimmelpenning-Feuerstein-Mims syndrome, which presents with neurologic, skeletal, genitourinary, cardiovascular, and ophthalmic disorders, in addition to cutaneous features. Others include phacomatosis pigmentokeratotica, didmyosis aplasticosebacea, SCALP syndrome (sebaceus nevus, central nervous system malformations, aplasia cutis congenita, limbal dermoid, and pigmented nevus), and more.4,5

Etiology
The etiology of NS has not been completely determined. One study that evaluated 44 NS tissue samples suggested the presence of human papillomavirus (HPV) in NS formation, finding that 82% of NS lesions studied contained HPV DNA. From these results, Carlson et al6 suggested a possible maternal transmission of HPV and infection of ectodermal cells as a potential cause of NS; however, this hypothesis was soon challenged by a study that showed a complete absence of HPV in 16 samples via histological evaluation and polymerase chain reaction for a broad range of HPV types.7 There were investigations into a patched (PTCH) deletion as the cause of NS and thus explained the historically high rate of secondary BCC.8 Further studies showed no mutations at the PTCH locus in trichoblastomas or other tumors arising from NS.9,10

More recent studies have recognized HRAS and KRAS mutations as a causative factor in NS.11 Nevus sebaceus belongs to a group of syndromes resulting from lethal mutations that survive via mosaicism. Nevus sebaceus is caused by postzygotic HRAS or KRAS mutations and is known as a mosaic RASopathy.12 In fact, there is growing evidence to suggest that other nevoid proliferations including keratinocytic epidermal nevi and melanocytic nevi also fall into the spectrum of mosaic RASopathies.13

Staging
There are 3 clinical stages of NS, originally described by Mehregan and Pinkus.14 In stage I (historically known as the infantile stage), the lesion presents as a yellow to pink, smooth, hairless patch. Histologic features include immature hair follicles and hypoplastic sebaceous glands. In stage II (also known as the puberty stage), the lesion becomes more pronounced. Firmer plaques can develop with hyperkeratosis. Hormonal changes cause sebaceous glands to develop, accompanied by epidermal hyperplasia and maturation of apocrine glands. Stage III (the tumoral stage) is a period that various neoplasms have the highest likelihood of occurring. Nevus sebaceus in an adolescent or adult demonstrates mature adnexal structures and greater epidermal hyperplasia.2,4,15

Malignancy
By virtue of these stages of NS development, malignant transformation is expected most often during stage III. However, cases have been reported of malignant tumor development in NS in children before puberty. Two case reports described a 7-year-old boy and a 10-year-old boy diagnosed with a BCC arising from an NS.16,17 However, secondary BCC formation before 16 years of age is rare. Basal cell carcinoma arising from an NS has been commonly reported and is the most common malignant neoplasm in NS (1.1%).2,3 However, the most common neoplasm overall is trichoblastoma (7.4%). The second most common tumor was syringocystadenoma papilliferum, occurring in approximately 5.2% of NS cases. The neoplasm rate in NS was found to be proportional to the patient age.2,18 Multiple studies have shown the overall rate of secondary neoplasms in NS to be 13% to 21.4%, with malignant tumors composing 0.8% to 2.5%.2,15,19 Other neoplasms that have been reported include keratoacanthoma, trichilemmoma, sebaceoma, nevocellular nevus, squamous cell carcinoma, adnexal carcinoma, apocrine adenocarcinoma, and malignant melanoma.19-21

It is argued that the reported rate of BCC formation is overestimated, as prior studies incorrectly labeled trichoblastomas as BCCs. In fact, the largest studies of NS from the 1990s revealed lower rates of malignant secondary tumors than previously determined.4

The identification of apocrine adenocarcinoma tumors arising from NS is exceedingly rare. A study performed by Cribier et al19 in 2000 retrospectively analyzed 596 cases of excised NS from 1932 to 1998. No apocrine carcinomas were reported in this study.19 Approximately 12 cases have been previously reported throughout the literature.20-26 Apocrine carcinomas occur most frequently in apocrine-rich areas such as the axillae, external ears, eyelids, and anogenital area. However, in the cases with apocrine carcinomas that developed from NS, the carcinomas have been located almost exclusively on the scalp.23

Histopathology
Histopathologic examination reveals considerable variation in morphology, and an underlying pattern has been difficult to recognize. Unfortunately, some authors have concluded that the diagnosis of apocrine carcinoma is relatively subjective.26 Robson et al26 identified 3 general architectural patterns: tubular, tubulopapillary, and solid. Tubular structures consisted of glands and ducts lined by a single or multilayered epithelium. Tubulopapillary architecture was characterized by epithelium forming papillary folds without a fibrovascular core. The solid morphology showed sheets of cells with limited ductal or tubular formation.26 The most specific criteria of these apocrine carcinomas are identification of decapitation secretion, periodic acid–Schiff–positive diastase-resistant material present in the cells or lumen, and positive immunostaining for gross cystic disease fluid protein-15.27

Robson et al26 reported estrogen receptor positivity and androgen receptor positivity in 62% and 64% of 24 primary apocrine carcinoma cases, respectively. However, whether these markers are as common in NS-related apocrine carcinomas has yet to be noted in the literature. One study reports a case of apocrine carcinoma from NS with positive staining for human epidermal growth factor-2, a cell membrane receptor tyrosine kinase commonly investigated in breast cancers and extramammary Paget disease.22

These apocrine carcinomas do have the potential for lymphatic metastasis, as seen with multiple studies. Domingo and Helwig21 identified regional lymph node metastasis in 2 of its 4 apocrine carcinoma patients. Robson et al26 reported lymphovascular invasion in 4 cases and perineural invasion in 2 of 24 patients studied. However, even in the context of recurrence and regional metastasis, the prognosis was good and seldom fatal.26

Treatment
The most effective treatment of NS is excision of dermal and epidermal components. Excision should be completed with a minimum of 2- to 3-mm margins and full thickness down to the underlying supporting fat.28 Historically, the practice of prophylactic excision of NS was supported by the potential for malignant transformation; however, early excision of NS may be less reasonable in light of these more recent studies showing lower incidence of BCC (0.8%), replaced by benign trichoblastomas.19 In the case of apocrine carcinoma development, excision is undoubtedly recommended, with unclear recommendations regarding further evaluation for metastasis.

Excision also may be favored for cosmetic purposes, given the visible regions where NS tends to develop. Chepla and Gosain29 argued that surgical intervention should be based on other factors such as location on the scalp, alopecia, and other issues affecting appearance and monitoring rather than incidence of malignant transformation. Close monitoring and biopsy of suspicious areas is a more conservative option.

Other therapies include CO2 laser, as demonstrated by Kiedrowicz et al,30 on linear NS in a patient with Schimmelpenning-Feuerstein-Mims syndrome.31 However, this approach is palliative and not effective in removing the entire lesion. Electrodesiccation and curettage and dermabrasion also are not good options for the same reason.4

Occurrence in Children
Nevus sebaceus in children, accompanied by other findings suggestive of epidermal nevus syndromes, should prompt further investigation. Schimmelpenning-Feuerstein-Mims syndrome includes major neurological abnormalities including hemimegalencephaly and seizures.32

Conclusion

Apocrine carcinomas are malignant neoplasms that may rarely arise within an NS. Their clinical identification is difficult and requires histopathologic evaluation. Upon recognition, prompt excision with tumor-free margins is recommended. As a rare entity, little data is available regarding its metastatic potential or overall survival rates. Further investigation is clearly necessary as new cases arise.

Nevus sebaceus (NS) is a benign hair follicle neoplasm present in approximately 1.3% of the population, typically involving the scalp, neck, or face.1 These lesions usually are present at birth or identified soon after, during the first year. They present as a yellowish hairless patch or plaque but can develop a more papillomatous appearance, especially after puberty. Historically, the concern with NS was its tendency to transform into basal cell carcinoma (BCC), which prompted surgical excision of the lesion during childhood. This theory has been discounted more recently, as further research has suggested that what was once thought to be BCC may have been confused with the similarly appearing trichoblastoma; however, malignant transformation of NS does still occur, with BCC still being the most common.2 We present the case of a long-standing NS with rare transformation to apocrine carcinoma.

Case Report

A 76-year-old woman presented with several new lesions within a previously diagnosed NS. She reported having the large plaque for as long as she could recall but reported that several new growths developed within the plaque over the last 2 months, slowly increasing in size. She reported a prior biopsy within the growth several years prior, which she described as an irritated seborrheic keratosis.

Physical examination demonstrated 4 distinct lesions within the flesh-colored, verrucous plaque located on the left side of the temporal scalp (Figure 1). The first lesion was a 2.5-cm pearly, pink, exophytic tumor (labeled as A in Figure 1). The next 2 lesions were brown, pedunculated, verrucous papules (labeled as B and C in Figure 1). The last lesion was a purple papule (labeled as D in Figure 1). Four shave biopsies were performed for histologic analysis of the lesions. Lesions B, C, and D were consistent with trichoblastomas, as pathology showed basaloid epithelial tumors that displayed primitive follicular structures, areas of stromal induction, and some pigmentation. Lesion A, originally thought to be suspicious for a BCC, was determined to be a primary cutaneous apocrine adenocarcinoma upon pathologic review. The pathology showed a dermal tumor displaying solid and tubular areas with decapitation secretion. Nuclear pleomorphism and mitoses were present (Figure 2), and staining for carcinoembryonic antigen was positive (Figure 3). Immunoreactivity with epithelial membrane antigen and cytokeratin 7 was noted as well as focal positivity for mammaglobin. Primary apocrine carcinoma was favored over metastatic carcinoma due to the location of the lesion within an NS along with a negative history of internal malignancy. Dermatopathology recommended complete removal of all lesions within the NS.

Figure1
Figure 1. Verrucous plaque and 4 distinct neoplasms (labeled A–D) on the left side of the temporal scalp.

Figure2
Figure 2. Apocrine carcinoma histopathology demonstrating solid and tubular areas with decapitation secretion (A)(H&E). On high power, marked nuclear pleomorphism and increased mitotic activity were observed (B)(H&E).

Figure3
Figure 3. Apocrine carcinoma demonstrating carcinoembryonic antigen staining positivity.


Upon discussing biopsy results and recommendations with our patient, she agreed to undergo excision with intraoperative pathology by a plastic surgeon within our practice to ensure clear margins. The surgical defect following excision was sizeable and closed utilizing a rhomboid flap, full-thickness skin graft, and a split-thickness skin graft. At surgical follow-up, she was doing well and there have been no signs of local recurrence for 10 months since excision.

 

 

Comment

Presentation
Nevus sebaceus is the most common adnexal tumor and is classified as a benign congenital hair follicle tumor that is located most commonly on the scalp but also occurs on the face and neck.1 The lesions usually are present at birth but also can develop during the first year of life.2 Diagnosis may be later, during adolescence, when patients seek medical attention during the lesion’s rapid growth phase.1 Nevus sebaceus also is known as an organoid nevus because it may contain all components of the skin. It was originally identified by Jadassohn in 1895.3 It presents as a yellowish, smooth, hairless patch or plaque in prepubertal patients. During adolescence, the lesion typically becomes more yellowish, as well as papillomatous, scaly, or warty. The reported incidence of NS is 0.05% to 1% in dermatology patients.2

Differential
Nevus sebaceus also is a component of several syndromes that should be kept in mind, including Schimmelpenning-Feuerstein-Mims syndrome, which presents with neurologic, skeletal, genitourinary, cardiovascular, and ophthalmic disorders, in addition to cutaneous features. Others include phacomatosis pigmentokeratotica, didmyosis aplasticosebacea, SCALP syndrome (sebaceus nevus, central nervous system malformations, aplasia cutis congenita, limbal dermoid, and pigmented nevus), and more.4,5

Etiology
The etiology of NS has not been completely determined. One study that evaluated 44 NS tissue samples suggested the presence of human papillomavirus (HPV) in NS formation, finding that 82% of NS lesions studied contained HPV DNA. From these results, Carlson et al6 suggested a possible maternal transmission of HPV and infection of ectodermal cells as a potential cause of NS; however, this hypothesis was soon challenged by a study that showed a complete absence of HPV in 16 samples via histological evaluation and polymerase chain reaction for a broad range of HPV types.7 There were investigations into a patched (PTCH) deletion as the cause of NS and thus explained the historically high rate of secondary BCC.8 Further studies showed no mutations at the PTCH locus in trichoblastomas or other tumors arising from NS.9,10

More recent studies have recognized HRAS and KRAS mutations as a causative factor in NS.11 Nevus sebaceus belongs to a group of syndromes resulting from lethal mutations that survive via mosaicism. Nevus sebaceus is caused by postzygotic HRAS or KRAS mutations and is known as a mosaic RASopathy.12 In fact, there is growing evidence to suggest that other nevoid proliferations including keratinocytic epidermal nevi and melanocytic nevi also fall into the spectrum of mosaic RASopathies.13

Staging
There are 3 clinical stages of NS, originally described by Mehregan and Pinkus.14 In stage I (historically known as the infantile stage), the lesion presents as a yellow to pink, smooth, hairless patch. Histologic features include immature hair follicles and hypoplastic sebaceous glands. In stage II (also known as the puberty stage), the lesion becomes more pronounced. Firmer plaques can develop with hyperkeratosis. Hormonal changes cause sebaceous glands to develop, accompanied by epidermal hyperplasia and maturation of apocrine glands. Stage III (the tumoral stage) is a period that various neoplasms have the highest likelihood of occurring. Nevus sebaceus in an adolescent or adult demonstrates mature adnexal structures and greater epidermal hyperplasia.2,4,15

Malignancy
By virtue of these stages of NS development, malignant transformation is expected most often during stage III. However, cases have been reported of malignant tumor development in NS in children before puberty. Two case reports described a 7-year-old boy and a 10-year-old boy diagnosed with a BCC arising from an NS.16,17 However, secondary BCC formation before 16 years of age is rare. Basal cell carcinoma arising from an NS has been commonly reported and is the most common malignant neoplasm in NS (1.1%).2,3 However, the most common neoplasm overall is trichoblastoma (7.4%). The second most common tumor was syringocystadenoma papilliferum, occurring in approximately 5.2% of NS cases. The neoplasm rate in NS was found to be proportional to the patient age.2,18 Multiple studies have shown the overall rate of secondary neoplasms in NS to be 13% to 21.4%, with malignant tumors composing 0.8% to 2.5%.2,15,19 Other neoplasms that have been reported include keratoacanthoma, trichilemmoma, sebaceoma, nevocellular nevus, squamous cell carcinoma, adnexal carcinoma, apocrine adenocarcinoma, and malignant melanoma.19-21

It is argued that the reported rate of BCC formation is overestimated, as prior studies incorrectly labeled trichoblastomas as BCCs. In fact, the largest studies of NS from the 1990s revealed lower rates of malignant secondary tumors than previously determined.4

The identification of apocrine adenocarcinoma tumors arising from NS is exceedingly rare. A study performed by Cribier et al19 in 2000 retrospectively analyzed 596 cases of excised NS from 1932 to 1998. No apocrine carcinomas were reported in this study.19 Approximately 12 cases have been previously reported throughout the literature.20-26 Apocrine carcinomas occur most frequently in apocrine-rich areas such as the axillae, external ears, eyelids, and anogenital area. However, in the cases with apocrine carcinomas that developed from NS, the carcinomas have been located almost exclusively on the scalp.23

Histopathology
Histopathologic examination reveals considerable variation in morphology, and an underlying pattern has been difficult to recognize. Unfortunately, some authors have concluded that the diagnosis of apocrine carcinoma is relatively subjective.26 Robson et al26 identified 3 general architectural patterns: tubular, tubulopapillary, and solid. Tubular structures consisted of glands and ducts lined by a single or multilayered epithelium. Tubulopapillary architecture was characterized by epithelium forming papillary folds without a fibrovascular core. The solid morphology showed sheets of cells with limited ductal or tubular formation.26 The most specific criteria of these apocrine carcinomas are identification of decapitation secretion, periodic acid–Schiff–positive diastase-resistant material present in the cells or lumen, and positive immunostaining for gross cystic disease fluid protein-15.27

Robson et al26 reported estrogen receptor positivity and androgen receptor positivity in 62% and 64% of 24 primary apocrine carcinoma cases, respectively. However, whether these markers are as common in NS-related apocrine carcinomas has yet to be noted in the literature. One study reports a case of apocrine carcinoma from NS with positive staining for human epidermal growth factor-2, a cell membrane receptor tyrosine kinase commonly investigated in breast cancers and extramammary Paget disease.22

These apocrine carcinomas do have the potential for lymphatic metastasis, as seen with multiple studies. Domingo and Helwig21 identified regional lymph node metastasis in 2 of its 4 apocrine carcinoma patients. Robson et al26 reported lymphovascular invasion in 4 cases and perineural invasion in 2 of 24 patients studied. However, even in the context of recurrence and regional metastasis, the prognosis was good and seldom fatal.26

Treatment
The most effective treatment of NS is excision of dermal and epidermal components. Excision should be completed with a minimum of 2- to 3-mm margins and full thickness down to the underlying supporting fat.28 Historically, the practice of prophylactic excision of NS was supported by the potential for malignant transformation; however, early excision of NS may be less reasonable in light of these more recent studies showing lower incidence of BCC (0.8%), replaced by benign trichoblastomas.19 In the case of apocrine carcinoma development, excision is undoubtedly recommended, with unclear recommendations regarding further evaluation for metastasis.

Excision also may be favored for cosmetic purposes, given the visible regions where NS tends to develop. Chepla and Gosain29 argued that surgical intervention should be based on other factors such as location on the scalp, alopecia, and other issues affecting appearance and monitoring rather than incidence of malignant transformation. Close monitoring and biopsy of suspicious areas is a more conservative option.

Other therapies include CO2 laser, as demonstrated by Kiedrowicz et al,30 on linear NS in a patient with Schimmelpenning-Feuerstein-Mims syndrome.31 However, this approach is palliative and not effective in removing the entire lesion. Electrodesiccation and curettage and dermabrasion also are not good options for the same reason.4

Occurrence in Children
Nevus sebaceus in children, accompanied by other findings suggestive of epidermal nevus syndromes, should prompt further investigation. Schimmelpenning-Feuerstein-Mims syndrome includes major neurological abnormalities including hemimegalencephaly and seizures.32

Conclusion

Apocrine carcinomas are malignant neoplasms that may rarely arise within an NS. Their clinical identification is difficult and requires histopathologic evaluation. Upon recognition, prompt excision with tumor-free margins is recommended. As a rare entity, little data is available regarding its metastatic potential or overall survival rates. Further investigation is clearly necessary as new cases arise.

References
  1. Kamyab-Hesari K, Balochi K, Afshar N, et al. Clinicopathological study of 1016 consecutive adnexal skin tumors. Acta Med Iran. 2013;51:879-885.
  2. Idriss MH, Elston DM. Secondary neoplasms associated with nevus sebaceus of Jadassohn: a study of 707 cases. J Am Acad Dermatol. 2014;70:332-337.
  3. Ball EA, Hussain M, Moss AL. Squamous cell carcinoma and basal cell carcinoma arising in a naevus sebaceous of Jadassohn: case report and literature review. Clin Exp Dermatol. 2005;30:259-260.
  4. Moody MN, Landau JM, Goldberg LH. Nevus sebaceous revisited. Pediatr Dermatol. 2012;29:15-23.
  5. Happle R. The group of epidermal nevus syndromes part I. well defined phenotypes. J Am Acad Dermatol. 2010;63:1-22; quiz 23-24.
  6. Carlson JA, Cribier B, Nuovo G, et al. Epidermodysplasia verruciformis-associated and genital-mucosal high-risk human papillomavirus DNA are prevalent in nevus sebaceus of Jadassohn. J Am Acad Dermatol. 2008;59:279-294.
  7. Kim D, Benjamin LT, Sahoo MK, et al. Human papilloma virus is not prevalent in nevus sebaceus [published online November 14, 2013]. Pediatr Dermatol. 2014;31:326-330.
  8. Xin H, Matt D, Qin JZ, et al. The sebaceous nevus: a nevus with deletions of the PTCH gene. Cancer Res. 1999;59:1834-1836.
  9. Hafner C, Schmiemann V, Ruetten A, et al. PTCH mutations are not mainly involved in the pathogenesis of sporadic trichoblastomas. Hum Pathol. 2007;38:1496-1500.
  10. Takata M, Tojo M, Hatta N, et al. No evidence of deregulated patched-hedgehog signaling pathway in trichoblastomas and other tumors arising within nevus sebaceous. J Invest Dermatol. 2001;117:1666-1670.
  11. Levinsohn JL, Tian LC, Boyden LM, et al. Whole-exome sequencing reveals somatic mutations in HRAS and KRAS, which cause nevus sebaceus [published online October 25, 2012]. J Invest Dermatol. 2013;133:827-830.
  12. Happle R. Nevus sebaceus is a mosaic RASopathy. J Invest Dermatol. 2013;133:597-600.
  13. Luo S, Tsao H. Epidermal, sebaceous, and melanocytic nevoid proliferations are spectrums of mosaic RASopathies. J Invest Dermatol. 2014;134:2493-2496.
  14. Mehregan AH, Pinkus H. Life history of organoid nevi. special reference to nevus sebaceus of Jadassohn. Arch Dermatol. 1965;91:574-588.
  15. Muñoz-Pérez MA, García-Hernandez MJ, Ríos JJ, et al. Sebaceus naevi: a clinicopathologic study. J Eur Acad Dermatol Venereol. 2002;16:319-324.
  16. Altaykan A, Ersoy-Evans S, Erkin G, et al. Basal cell carcinoma arising in nevus sebaceous during childhood. Pediatr Dermatol. 2008;25:616-619.
  17. Turner CD, Shea CR, Rosoff PM. Basal cell carcinoma originating from a nevus sebaceus on the scalp of a 7-year-old boy. J Pediatr Hematol Oncol. 2001;23:247-249.
  18. Jaqueti G, Requena L, Sánchez Yus E. Trichoblastoma is the most common neoplasm developed in nevus sebaceus of Jadassohn: a clinicopathologic study of a series of 155 cases. Am J Dermatopathol. 2000;22:108-118.
  19. Cribier B, Scrivener Y, Grosshans E. Tumors arising in nevus sebaceus: a study of 596 cases. J Am Acad Dermatol. 2000;42(2, pt 1):263-268.
  20. Paudel U, Jha A, Pokhrel DB, et al. Apocrine carcinoma developing in a naevus sebaceous of scalp. Kathmandu Univ Med J (KUMJ). 2012;10:103-105.
  21. Domingo J, Helwig EB. Malignant neoplasms associated with nevus sebaceus of Jadassohn. J Am Acad Dermatol. 1979;1:545-556.
  22. Tanese K, Wakabayashi A, Suzuki T, et al. Immunoexpression of human epidermal growth factor receptor-2 in apocrine carcinoma arising in naevus sebaceous, case report [published online August 23, 2009]. J Eur Acad Dermatol Venereol. 2010;24:360-362.
  23. Dalle S, Skowron F, Balme B, et al. Apocrine carcinoma developed in nevus sebaceus of Jadassohn. Eur J Dermatol. 2003;13:487-489.
  24. Jacyk WK, Requena L, Sánchez Yus E, et al. Tubular apocrine carcinoma arising in a nevus sebaceus of Jadassohn. Am J Dermatopathol. 1998;20:389-392.
  25. Ansai S, Koseki S, Hashimoto H, et al. A case of ductal sweat gland carcinoma connected to syringocystadenoma papilliferum arising in nevus sebaceus. J Cutan Pathol. 1994;21:557-563.
  26. Robson A, Lazar AJ, Ben Nagi J, et al. Primary cutaneous apocrine carcinoma: a clinico-pathologic analysis of 24 cases. Am J Surg Pathol. 2008;32:682-690.
  27. Paties C, Taccagni GL, Papotti M, et al. Apocrine carcinoma of the skin. a clinicopathologic, immunocytochemical, and ultrastructural study. Cancer. 1993;71:375-381.
  28. Davison SP, Khachemoune A, Yu D, et al. Nevus sebaceus of Jadassohn revisited with reconstruction options. Int J Dermatol. 2005;44:145-150.
  29. Chepla KJ, Gosain AK. Giant nevus sebaceus: definition, surgical techniques, and rationale for treatment. Plast Reconstr Surg. 2012;130:296E-304E.
  30. Kiedrowicz M, Kacalak-Rzepka A, Królicki A et al. Therapeutic effects of CO2 laser therapy of linear nevus sebaceous in the course of the Schimmelpenning-Feuerstein-Mims syndrome. Postepy Dermatol Allergol. 2013;30:320-323.
  31. Ashinoff R. Linear nevus sebaceus of Jadassohn treated with the carbon dioxide laser. Pediatr Dermatol. 1993;10:189-191.
  32. van de Warrenburg BP, van Gulik S, Renier WO, et al. The linear naevus sebaceus syndrome. Clin Neurol Neurosurg. 1998;100:126-132.
References
  1. Kamyab-Hesari K, Balochi K, Afshar N, et al. Clinicopathological study of 1016 consecutive adnexal skin tumors. Acta Med Iran. 2013;51:879-885.
  2. Idriss MH, Elston DM. Secondary neoplasms associated with nevus sebaceus of Jadassohn: a study of 707 cases. J Am Acad Dermatol. 2014;70:332-337.
  3. Ball EA, Hussain M, Moss AL. Squamous cell carcinoma and basal cell carcinoma arising in a naevus sebaceous of Jadassohn: case report and literature review. Clin Exp Dermatol. 2005;30:259-260.
  4. Moody MN, Landau JM, Goldberg LH. Nevus sebaceous revisited. Pediatr Dermatol. 2012;29:15-23.
  5. Happle R. The group of epidermal nevus syndromes part I. well defined phenotypes. J Am Acad Dermatol. 2010;63:1-22; quiz 23-24.
  6. Carlson JA, Cribier B, Nuovo G, et al. Epidermodysplasia verruciformis-associated and genital-mucosal high-risk human papillomavirus DNA are prevalent in nevus sebaceus of Jadassohn. J Am Acad Dermatol. 2008;59:279-294.
  7. Kim D, Benjamin LT, Sahoo MK, et al. Human papilloma virus is not prevalent in nevus sebaceus [published online November 14, 2013]. Pediatr Dermatol. 2014;31:326-330.
  8. Xin H, Matt D, Qin JZ, et al. The sebaceous nevus: a nevus with deletions of the PTCH gene. Cancer Res. 1999;59:1834-1836.
  9. Hafner C, Schmiemann V, Ruetten A, et al. PTCH mutations are not mainly involved in the pathogenesis of sporadic trichoblastomas. Hum Pathol. 2007;38:1496-1500.
  10. Takata M, Tojo M, Hatta N, et al. No evidence of deregulated patched-hedgehog signaling pathway in trichoblastomas and other tumors arising within nevus sebaceous. J Invest Dermatol. 2001;117:1666-1670.
  11. Levinsohn JL, Tian LC, Boyden LM, et al. Whole-exome sequencing reveals somatic mutations in HRAS and KRAS, which cause nevus sebaceus [published online October 25, 2012]. J Invest Dermatol. 2013;133:827-830.
  12. Happle R. Nevus sebaceus is a mosaic RASopathy. J Invest Dermatol. 2013;133:597-600.
  13. Luo S, Tsao H. Epidermal, sebaceous, and melanocytic nevoid proliferations are spectrums of mosaic RASopathies. J Invest Dermatol. 2014;134:2493-2496.
  14. Mehregan AH, Pinkus H. Life history of organoid nevi. special reference to nevus sebaceus of Jadassohn. Arch Dermatol. 1965;91:574-588.
  15. Muñoz-Pérez MA, García-Hernandez MJ, Ríos JJ, et al. Sebaceus naevi: a clinicopathologic study. J Eur Acad Dermatol Venereol. 2002;16:319-324.
  16. Altaykan A, Ersoy-Evans S, Erkin G, et al. Basal cell carcinoma arising in nevus sebaceous during childhood. Pediatr Dermatol. 2008;25:616-619.
  17. Turner CD, Shea CR, Rosoff PM. Basal cell carcinoma originating from a nevus sebaceus on the scalp of a 7-year-old boy. J Pediatr Hematol Oncol. 2001;23:247-249.
  18. Jaqueti G, Requena L, Sánchez Yus E. Trichoblastoma is the most common neoplasm developed in nevus sebaceus of Jadassohn: a clinicopathologic study of a series of 155 cases. Am J Dermatopathol. 2000;22:108-118.
  19. Cribier B, Scrivener Y, Grosshans E. Tumors arising in nevus sebaceus: a study of 596 cases. J Am Acad Dermatol. 2000;42(2, pt 1):263-268.
  20. Paudel U, Jha A, Pokhrel DB, et al. Apocrine carcinoma developing in a naevus sebaceous of scalp. Kathmandu Univ Med J (KUMJ). 2012;10:103-105.
  21. Domingo J, Helwig EB. Malignant neoplasms associated with nevus sebaceus of Jadassohn. J Am Acad Dermatol. 1979;1:545-556.
  22. Tanese K, Wakabayashi A, Suzuki T, et al. Immunoexpression of human epidermal growth factor receptor-2 in apocrine carcinoma arising in naevus sebaceous, case report [published online August 23, 2009]. J Eur Acad Dermatol Venereol. 2010;24:360-362.
  23. Dalle S, Skowron F, Balme B, et al. Apocrine carcinoma developed in nevus sebaceus of Jadassohn. Eur J Dermatol. 2003;13:487-489.
  24. Jacyk WK, Requena L, Sánchez Yus E, et al. Tubular apocrine carcinoma arising in a nevus sebaceus of Jadassohn. Am J Dermatopathol. 1998;20:389-392.
  25. Ansai S, Koseki S, Hashimoto H, et al. A case of ductal sweat gland carcinoma connected to syringocystadenoma papilliferum arising in nevus sebaceus. J Cutan Pathol. 1994;21:557-563.
  26. Robson A, Lazar AJ, Ben Nagi J, et al. Primary cutaneous apocrine carcinoma: a clinico-pathologic analysis of 24 cases. Am J Surg Pathol. 2008;32:682-690.
  27. Paties C, Taccagni GL, Papotti M, et al. Apocrine carcinoma of the skin. a clinicopathologic, immunocytochemical, and ultrastructural study. Cancer. 1993;71:375-381.
  28. Davison SP, Khachemoune A, Yu D, et al. Nevus sebaceus of Jadassohn revisited with reconstruction options. Int J Dermatol. 2005;44:145-150.
  29. Chepla KJ, Gosain AK. Giant nevus sebaceus: definition, surgical techniques, and rationale for treatment. Plast Reconstr Surg. 2012;130:296E-304E.
  30. Kiedrowicz M, Kacalak-Rzepka A, Królicki A et al. Therapeutic effects of CO2 laser therapy of linear nevus sebaceous in the course of the Schimmelpenning-Feuerstein-Mims syndrome. Postepy Dermatol Allergol. 2013;30:320-323.
  31. Ashinoff R. Linear nevus sebaceus of Jadassohn treated with the carbon dioxide laser. Pediatr Dermatol. 1993;10:189-191.
  32. van de Warrenburg BP, van Gulik S, Renier WO, et al. The linear naevus sebaceus syndrome. Clin Neurol Neurosurg. 1998;100:126-132.
Issue
Cutis - 102(4)
Issue
Cutis - 102(4)
Page Number
291-294
Page Number
291-294
Publications
Publications
Topics
Article Type
Sections
Inside the Article

Practice Points

  • Nevus sebaceus (NS) in the centrofacial region has been correlated with a higher risk for neurological abnormalities, including intellectual disability and seizures.
  • Historically, basal cell carcinomas (BCCs) were considered a common occurrence arising from an NS, prompting prophylactic surgical excision of such lesions.
  • More recently, it has been recognized that the most common tumor to arise from NS is trichoblastoma rather than BCC; in fact, BCC and other malignancies have been found to be relatively rare compared to their benign counterparts.
  • In light of this discovery, observation of NS may be a more prudent course of treatment versus prophylactic surgical excision.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Article PDF Media

Leukemia Cutis in Acute Myeloid Leukemia Signifies a Poor Prognosis

Article Type
Changed
Thu, 01/10/2019 - 13:54
Display Headline
Leukemia Cutis in Acute Myeloid Leukemia Signifies a Poor Prognosis

Case Report

A 66-year-old man with a history of type 2 diabetes mellitus presented with considerable muscle weakness and infiltrative, flesh-colored plaques on the face, trunk, and arms of 3 months’ duration. The patient required the use of a wheelchair due to muscle weakness. On physical examination he had diffuse, infiltrative, flesh-colored plaques on the entire face (Figure 1A), trunk, and arms. The eyelids and lips were swollen, and the nose was distorted due to the infiltrative plaques (Figure 1B). Additionally, there were hypopigmented macules and patches scattered among the infiltrative plaques on the face, trunk, and arms (Figure 1C).

Figure1
Figure 1. Leukemia cutis presenting as diffuse, infiltrative, flesh-colored plaques on the face (A). The eyelids and lips were swollen and the nose was distorted due to the infiltrative plaques (B). Hypopigmented macules and patches were scattered among the infiltrative plaques of leukemia cutis on the left arm (C).

Punch biopsy specimens were obtained from the left cheek and left upper arm and were submitted for histologic examination with routine hematoxylin and eosin staining (Figure 2). Histopathology showed infiltrating and diffuse monomorphic cells in the dermis with large and hyperchromatic nuclei. Some nuclei were cleaved or folded in configuration. The cells displayed ample surrounding cytoplasm, which was finely granular or vacuolated. The infiltrate was accentuated in the perifollicular adventitial dermis. Immunohistochemistry was positive for CD33 and negative for CD3, CD20, and myeloperoxidase. Additionally, periodic acid–Schiff and Fite stains were negative for microorganisms. These morphologic and immunohistochemical findings were consistent with acute myeloid leukemia (AML). Further testing with complete blood cell count, peripheral blood smear, and bone marrow biopsy confirmed the diagnosis of AML. The patient subsequently died 5 weeks later.

Figure 2. Punch biopsy from the left cheek (A) and left upper arm (B) showed positive staining for acute myeloid leukemia (H&E, original magnifications ×200 and ×100). Specifically, monomorphic cells with large hyperchromatic nuclei were observed infiltrating the dermis, occasionally lining single file between collagen bundles. The cells stained positive for CD33 and negative for CD3 and CD20.

Comment

Presentation of LC
Thirty percent to 40% of leukemia patients present with a variety of nonspecific cutaneous signs, including those related to hemorrhage, infection, and drug eruptions, as well as paraneoplastic lesions.1 Cutaneous signs of leukemia are less commonly due to leukemia cutis (LC), defined as the neoplastic infiltration of the skin or subcutaneous tissue by leukemic cells. The clinical presentation of LC varies, making it difficult to diagnose without immunohistochemistry. It can pre-sent as single or multiple erythematous papules and/or nodules, infiltrated plaques, macules, palpable purpura, ulcers, ecchymoses, and/or vesicles.2 Leukemia cutis most often presents on the head, neck, trunk, and sites of current or prior trauma. Gingival hyperplasia is another associated finding in the acute monocytic and myelomonocytic types of AML.3 Additionally, chloromas or granulocytic sarcomas are dermal nodules that can pre-sent in myelogenous leukemia.4

LC and AML
Leukemia cutis most commonly is observed in AML compared to the other types of leukemia. The myelomonocytic and monocytic subtypes of AML are most often implicated.5,6 The majority of patients with LC present with a pre-established (55%–77%) or simultaneous diagnosis of systemic leukemia (23%–38%). Rarely do patients present with LC with lack of systemic involvement and a normal peripheral smear (7%),2 which would be diagnosed as aleukemic leukemia.2,7 Furthermore, LC highly correlates with sites of additional extramedullary involvement; thus, the presence of LC in AML often signifies a poor prognosis.8 The 2-year survival rate for AML patients without LC is 30%, but for AML patients with LC it is only 6%.1

Histopathology
In LC, histology typically reveals a normal epidermis and nodular or diffuse infiltrating cells in the dermis. The cells can appear monomorphic, atypical, or immature, and there is occasional single-filing between collagen bundles. Causative types of neoplasms can be distinguished based on their morphologic, immunophenotypic, and cytogenetic properties.8-10

Incidence
Of the acute leukemias, AML accounts for the highest prevalence in adults,11 with an annual incidence of 14,590 cases in the United States.12 The incidence of AML increases with age; the mean age of patients diagnosed with AML is 67 years.12 Risk is increased with a history of exposure to radiotherapy, chemotherapy, or cigarette smoke; preexisting myeloproliferative or myelodysplastic syndromes and mutations in DNA repair (eg, Fanconi anemia); neutropenia (eg, from elastase mutations); and Down syndrome.13

Diagnosis
More than 20% blasts in the bone marrow is required for a diagnosis of AML.14 Specific to AML is the presence of large immature precursor cells with a granular cytoplasm and, when present, highly diagnostic Auer rods.12Acute myeloid leukemia can be distinguished by staining for myeloperoxidase; Sudan Black B; or the antigens CD13, CD33, or c-kit.15

In our case, CD33 was positive, which is a characteristic finding in AML. Myeloperoxidase also can be positive in AML; however, in our case it was negative, and it can be an insensitive marker in the context of LC. Although most cases of LC present concurrently with bone marrow infiltration, some cases present before systemic involvement; for example, granulocytic sarcomas can occur months earlier than the development of systemic leukemia. Thus, early detection by a dermatologist is essential. Depending on the lesion’s appearance, the differential diagnoses can include lymphoma, drug eruptions, infectious etiologies, sarcoidosis, metastases from other malignancies, and blistering dermatoses.

Management
Systemic therapy should be the cornerstone of therapy. Induction therapy includes the combined use of cytarabine (except in acute promyelocytic leukemia [M3], for which all-trans retinoic acid is indicated) and anthracycline derivatives in a “7+3” regimen to achieve complete remission. Specifically, cytarabine (100–200 mg/m2) typically is continuously administered intravenously for 7 days combined with intravenous administration of either daunorubicin (60–90 mg/m2) or idarubicin (12 mg/m2) on days 1, 2, and 3. Postremission therapy is highly individualized depending on patients’ prognostic factors and is indicated to reduce the likelihood of relapse and to improve patient mortality. High doses of cytarabine and hematopoietic stem cell transplantation commonly are utilized.12 Resolution of hematologic atypia may result in complete or partial resolution of LC.10

Conclusion

We diagnosed AML with systemic involvement in our patient based on the cutaneous manifestation of LC. Diagnosis of LC relies on immunohistochemistry and strong clinical suspicion, as cutaneous findings are diverse and nonspecific. Early recognition is essential, as LC in the context of systemic involvement portends a poor prognosis. Our patient died 5 weeks following initial presentation.

References
  1. Rao AG, Danturty I. Leukemia cutis. Indian J Dermatol. 2012;57:504.
  2. Su WPD, Buechner SA, Chin-Yang L. Clinicopathologic correlations in leukemia cutis. J Am Acad Dermatol. 1984;11:121-128.
  3. Kumar M, Nair V, Mishra L, et al. Gingival hyperplasia—a clue to the diagnosis of acute leukemia? Arch Oral Sci Res. 2012;2:165-168.
  4. Winfield H, Smoller B. Other lymphoproliferative and myeloproliferative diseases. In: Bolognia JL, Jorizzo JL, Schaffer JV, eds. Dermatology. 3rd ed. Philadelphia, PA: Mosby/Elsevier; 2012:2037-2048.
  5. Babina T, Miller L, Thomas B. Leukemia cutis. J Drugs Dermatol. 2012;11:416-417.
  6. Tziotzios C, Makrygeorgou A. Leukemia cutis. Cleve Clin J Med. 2011;78:226-227.
  7. Ratnam KV, Khor CJL, Su WPD. Leukemia cutis. Dermatol Clin 1994;12:419-431.
  8. Cho-Vega JH, Medeiros LJ, Prieto VG, et al. Leukemia cutis. Am J Clin Pathol. 2008;129:130-142.
  9. Buechner SA, Li CY, Su WP. Leukemia cutis. a histopathologic study of 42 cases. Am J Dermatopathol. 1985;7:109-119.
  10. Wagner G, Fenchel K, Back W, et al. Leukemia cutis—epidemiology, clinical presentation, and differential diagnoses. J Dtsch Dermatol Ges. 2012;10:27-36.
  11. O’Donnell MR, Abboud CN, Altman J, et al. NCCN Clinical Practice Guidelines acute myeloid leukemia. J Natl Compr Canc Netw. 2012;10:984-1021.
  12. Marcucci G, Bloomfield CD. Acute myeloid leukemia. In: Kasper DL, Fauci AS, Hauser SL, et al, eds. Harrison’s Principles of Internal Medicine. 19th ed. New York, NY: McGraw-Hill; 2015:678-686.
  13. Aster JC, DeAngelo DJ. Acute leukemias. In: Bunn HF, Aster JC, eds. Pathophysiology of Blood Disorders. New York, NY: McGraw-Hill; 2010:244-259.
  14. Damon LE, Andreadis C. Blood disorders. In: Papadakis MA, McPhee SJ, Rabow MW, eds. Current Medical Diagnosis & Treatment 2016. New York, NY: McGraw-Hill; 2016:495-541.
  15. Parikh SA, Jabbour E, Koller CA. Adult acute myeloid leukemia. In: Kantarjian HM, Wolff RA, eds. The MD Anderson Manual of Medical Oncology. 2nd ed. New York, NY: McGraw-Hill; 2011:15-32.
Article PDF
Author and Disclosure Information

From the Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton. Dr. Weatherall is from the Department of Clinical Biomedical Science. Dr. Weatherall also is from ClearlyDerm, Boca Raton.

The authors report no conflict of interest.

Correspondence: Jolie A. Krooks, BS, 77 Glades Rd, Boca Raton, FL 33431 (jkrooks2014@health.fau.edu).

Issue
Cutis - 102(4)
Publications
Topics
Page Number
266, 271-272
Sections
Author and Disclosure Information

From the Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton. Dr. Weatherall is from the Department of Clinical Biomedical Science. Dr. Weatherall also is from ClearlyDerm, Boca Raton.

The authors report no conflict of interest.

Correspondence: Jolie A. Krooks, BS, 77 Glades Rd, Boca Raton, FL 33431 (jkrooks2014@health.fau.edu).

Author and Disclosure Information

From the Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton. Dr. Weatherall is from the Department of Clinical Biomedical Science. Dr. Weatherall also is from ClearlyDerm, Boca Raton.

The authors report no conflict of interest.

Correspondence: Jolie A. Krooks, BS, 77 Glades Rd, Boca Raton, FL 33431 (jkrooks2014@health.fau.edu).

Article PDF
Article PDF

Case Report

A 66-year-old man with a history of type 2 diabetes mellitus presented with considerable muscle weakness and infiltrative, flesh-colored plaques on the face, trunk, and arms of 3 months’ duration. The patient required the use of a wheelchair due to muscle weakness. On physical examination he had diffuse, infiltrative, flesh-colored plaques on the entire face (Figure 1A), trunk, and arms. The eyelids and lips were swollen, and the nose was distorted due to the infiltrative plaques (Figure 1B). Additionally, there were hypopigmented macules and patches scattered among the infiltrative plaques on the face, trunk, and arms (Figure 1C).

Figure1
Figure 1. Leukemia cutis presenting as diffuse, infiltrative, flesh-colored plaques on the face (A). The eyelids and lips were swollen and the nose was distorted due to the infiltrative plaques (B). Hypopigmented macules and patches were scattered among the infiltrative plaques of leukemia cutis on the left arm (C).

Punch biopsy specimens were obtained from the left cheek and left upper arm and were submitted for histologic examination with routine hematoxylin and eosin staining (Figure 2). Histopathology showed infiltrating and diffuse monomorphic cells in the dermis with large and hyperchromatic nuclei. Some nuclei were cleaved or folded in configuration. The cells displayed ample surrounding cytoplasm, which was finely granular or vacuolated. The infiltrate was accentuated in the perifollicular adventitial dermis. Immunohistochemistry was positive for CD33 and negative for CD3, CD20, and myeloperoxidase. Additionally, periodic acid–Schiff and Fite stains were negative for microorganisms. These morphologic and immunohistochemical findings were consistent with acute myeloid leukemia (AML). Further testing with complete blood cell count, peripheral blood smear, and bone marrow biopsy confirmed the diagnosis of AML. The patient subsequently died 5 weeks later.

Figure 2. Punch biopsy from the left cheek (A) and left upper arm (B) showed positive staining for acute myeloid leukemia (H&E, original magnifications ×200 and ×100). Specifically, monomorphic cells with large hyperchromatic nuclei were observed infiltrating the dermis, occasionally lining single file between collagen bundles. The cells stained positive for CD33 and negative for CD3 and CD20.

Comment

Presentation of LC
Thirty percent to 40% of leukemia patients present with a variety of nonspecific cutaneous signs, including those related to hemorrhage, infection, and drug eruptions, as well as paraneoplastic lesions.1 Cutaneous signs of leukemia are less commonly due to leukemia cutis (LC), defined as the neoplastic infiltration of the skin or subcutaneous tissue by leukemic cells. The clinical presentation of LC varies, making it difficult to diagnose without immunohistochemistry. It can pre-sent as single or multiple erythematous papules and/or nodules, infiltrated plaques, macules, palpable purpura, ulcers, ecchymoses, and/or vesicles.2 Leukemia cutis most often presents on the head, neck, trunk, and sites of current or prior trauma. Gingival hyperplasia is another associated finding in the acute monocytic and myelomonocytic types of AML.3 Additionally, chloromas or granulocytic sarcomas are dermal nodules that can pre-sent in myelogenous leukemia.4

LC and AML
Leukemia cutis most commonly is observed in AML compared to the other types of leukemia. The myelomonocytic and monocytic subtypes of AML are most often implicated.5,6 The majority of patients with LC present with a pre-established (55%–77%) or simultaneous diagnosis of systemic leukemia (23%–38%). Rarely do patients present with LC with lack of systemic involvement and a normal peripheral smear (7%),2 which would be diagnosed as aleukemic leukemia.2,7 Furthermore, LC highly correlates with sites of additional extramedullary involvement; thus, the presence of LC in AML often signifies a poor prognosis.8 The 2-year survival rate for AML patients without LC is 30%, but for AML patients with LC it is only 6%.1

Histopathology
In LC, histology typically reveals a normal epidermis and nodular or diffuse infiltrating cells in the dermis. The cells can appear monomorphic, atypical, or immature, and there is occasional single-filing between collagen bundles. Causative types of neoplasms can be distinguished based on their morphologic, immunophenotypic, and cytogenetic properties.8-10

Incidence
Of the acute leukemias, AML accounts for the highest prevalence in adults,11 with an annual incidence of 14,590 cases in the United States.12 The incidence of AML increases with age; the mean age of patients diagnosed with AML is 67 years.12 Risk is increased with a history of exposure to radiotherapy, chemotherapy, or cigarette smoke; preexisting myeloproliferative or myelodysplastic syndromes and mutations in DNA repair (eg, Fanconi anemia); neutropenia (eg, from elastase mutations); and Down syndrome.13

Diagnosis
More than 20% blasts in the bone marrow is required for a diagnosis of AML.14 Specific to AML is the presence of large immature precursor cells with a granular cytoplasm and, when present, highly diagnostic Auer rods.12Acute myeloid leukemia can be distinguished by staining for myeloperoxidase; Sudan Black B; or the antigens CD13, CD33, or c-kit.15

In our case, CD33 was positive, which is a characteristic finding in AML. Myeloperoxidase also can be positive in AML; however, in our case it was negative, and it can be an insensitive marker in the context of LC. Although most cases of LC present concurrently with bone marrow infiltration, some cases present before systemic involvement; for example, granulocytic sarcomas can occur months earlier than the development of systemic leukemia. Thus, early detection by a dermatologist is essential. Depending on the lesion’s appearance, the differential diagnoses can include lymphoma, drug eruptions, infectious etiologies, sarcoidosis, metastases from other malignancies, and blistering dermatoses.

Management
Systemic therapy should be the cornerstone of therapy. Induction therapy includes the combined use of cytarabine (except in acute promyelocytic leukemia [M3], for which all-trans retinoic acid is indicated) and anthracycline derivatives in a “7+3” regimen to achieve complete remission. Specifically, cytarabine (100–200 mg/m2) typically is continuously administered intravenously for 7 days combined with intravenous administration of either daunorubicin (60–90 mg/m2) or idarubicin (12 mg/m2) on days 1, 2, and 3. Postremission therapy is highly individualized depending on patients’ prognostic factors and is indicated to reduce the likelihood of relapse and to improve patient mortality. High doses of cytarabine and hematopoietic stem cell transplantation commonly are utilized.12 Resolution of hematologic atypia may result in complete or partial resolution of LC.10

Conclusion

We diagnosed AML with systemic involvement in our patient based on the cutaneous manifestation of LC. Diagnosis of LC relies on immunohistochemistry and strong clinical suspicion, as cutaneous findings are diverse and nonspecific. Early recognition is essential, as LC in the context of systemic involvement portends a poor prognosis. Our patient died 5 weeks following initial presentation.

Case Report

A 66-year-old man with a history of type 2 diabetes mellitus presented with considerable muscle weakness and infiltrative, flesh-colored plaques on the face, trunk, and arms of 3 months’ duration. The patient required the use of a wheelchair due to muscle weakness. On physical examination he had diffuse, infiltrative, flesh-colored plaques on the entire face (Figure 1A), trunk, and arms. The eyelids and lips were swollen, and the nose was distorted due to the infiltrative plaques (Figure 1B). Additionally, there were hypopigmented macules and patches scattered among the infiltrative plaques on the face, trunk, and arms (Figure 1C).

Figure1
Figure 1. Leukemia cutis presenting as diffuse, infiltrative, flesh-colored plaques on the face (A). The eyelids and lips were swollen and the nose was distorted due to the infiltrative plaques (B). Hypopigmented macules and patches were scattered among the infiltrative plaques of leukemia cutis on the left arm (C).

Punch biopsy specimens were obtained from the left cheek and left upper arm and were submitted for histologic examination with routine hematoxylin and eosin staining (Figure 2). Histopathology showed infiltrating and diffuse monomorphic cells in the dermis with large and hyperchromatic nuclei. Some nuclei were cleaved or folded in configuration. The cells displayed ample surrounding cytoplasm, which was finely granular or vacuolated. The infiltrate was accentuated in the perifollicular adventitial dermis. Immunohistochemistry was positive for CD33 and negative for CD3, CD20, and myeloperoxidase. Additionally, periodic acid–Schiff and Fite stains were negative for microorganisms. These morphologic and immunohistochemical findings were consistent with acute myeloid leukemia (AML). Further testing with complete blood cell count, peripheral blood smear, and bone marrow biopsy confirmed the diagnosis of AML. The patient subsequently died 5 weeks later.

Figure 2. Punch biopsy from the left cheek (A) and left upper arm (B) showed positive staining for acute myeloid leukemia (H&E, original magnifications ×200 and ×100). Specifically, monomorphic cells with large hyperchromatic nuclei were observed infiltrating the dermis, occasionally lining single file between collagen bundles. The cells stained positive for CD33 and negative for CD3 and CD20.

Comment

Presentation of LC
Thirty percent to 40% of leukemia patients present with a variety of nonspecific cutaneous signs, including those related to hemorrhage, infection, and drug eruptions, as well as paraneoplastic lesions.1 Cutaneous signs of leukemia are less commonly due to leukemia cutis (LC), defined as the neoplastic infiltration of the skin or subcutaneous tissue by leukemic cells. The clinical presentation of LC varies, making it difficult to diagnose without immunohistochemistry. It can pre-sent as single or multiple erythematous papules and/or nodules, infiltrated plaques, macules, palpable purpura, ulcers, ecchymoses, and/or vesicles.2 Leukemia cutis most often presents on the head, neck, trunk, and sites of current or prior trauma. Gingival hyperplasia is another associated finding in the acute monocytic and myelomonocytic types of AML.3 Additionally, chloromas or granulocytic sarcomas are dermal nodules that can pre-sent in myelogenous leukemia.4

LC and AML
Leukemia cutis most commonly is observed in AML compared to the other types of leukemia. The myelomonocytic and monocytic subtypes of AML are most often implicated.5,6 The majority of patients with LC present with a pre-established (55%–77%) or simultaneous diagnosis of systemic leukemia (23%–38%). Rarely do patients present with LC with lack of systemic involvement and a normal peripheral smear (7%),2 which would be diagnosed as aleukemic leukemia.2,7 Furthermore, LC highly correlates with sites of additional extramedullary involvement; thus, the presence of LC in AML often signifies a poor prognosis.8 The 2-year survival rate for AML patients without LC is 30%, but for AML patients with LC it is only 6%.1

Histopathology
In LC, histology typically reveals a normal epidermis and nodular or diffuse infiltrating cells in the dermis. The cells can appear monomorphic, atypical, or immature, and there is occasional single-filing between collagen bundles. Causative types of neoplasms can be distinguished based on their morphologic, immunophenotypic, and cytogenetic properties.8-10

Incidence
Of the acute leukemias, AML accounts for the highest prevalence in adults,11 with an annual incidence of 14,590 cases in the United States.12 The incidence of AML increases with age; the mean age of patients diagnosed with AML is 67 years.12 Risk is increased with a history of exposure to radiotherapy, chemotherapy, or cigarette smoke; preexisting myeloproliferative or myelodysplastic syndromes and mutations in DNA repair (eg, Fanconi anemia); neutropenia (eg, from elastase mutations); and Down syndrome.13

Diagnosis
More than 20% blasts in the bone marrow is required for a diagnosis of AML.14 Specific to AML is the presence of large immature precursor cells with a granular cytoplasm and, when present, highly diagnostic Auer rods.12Acute myeloid leukemia can be distinguished by staining for myeloperoxidase; Sudan Black B; or the antigens CD13, CD33, or c-kit.15

In our case, CD33 was positive, which is a characteristic finding in AML. Myeloperoxidase also can be positive in AML; however, in our case it was negative, and it can be an insensitive marker in the context of LC. Although most cases of LC present concurrently with bone marrow infiltration, some cases present before systemic involvement; for example, granulocytic sarcomas can occur months earlier than the development of systemic leukemia. Thus, early detection by a dermatologist is essential. Depending on the lesion’s appearance, the differential diagnoses can include lymphoma, drug eruptions, infectious etiologies, sarcoidosis, metastases from other malignancies, and blistering dermatoses.

Management
Systemic therapy should be the cornerstone of therapy. Induction therapy includes the combined use of cytarabine (except in acute promyelocytic leukemia [M3], for which all-trans retinoic acid is indicated) and anthracycline derivatives in a “7+3” regimen to achieve complete remission. Specifically, cytarabine (100–200 mg/m2) typically is continuously administered intravenously for 7 days combined with intravenous administration of either daunorubicin (60–90 mg/m2) or idarubicin (12 mg/m2) on days 1, 2, and 3. Postremission therapy is highly individualized depending on patients’ prognostic factors and is indicated to reduce the likelihood of relapse and to improve patient mortality. High doses of cytarabine and hematopoietic stem cell transplantation commonly are utilized.12 Resolution of hematologic atypia may result in complete or partial resolution of LC.10

Conclusion

We diagnosed AML with systemic involvement in our patient based on the cutaneous manifestation of LC. Diagnosis of LC relies on immunohistochemistry and strong clinical suspicion, as cutaneous findings are diverse and nonspecific. Early recognition is essential, as LC in the context of systemic involvement portends a poor prognosis. Our patient died 5 weeks following initial presentation.

References
  1. Rao AG, Danturty I. Leukemia cutis. Indian J Dermatol. 2012;57:504.
  2. Su WPD, Buechner SA, Chin-Yang L. Clinicopathologic correlations in leukemia cutis. J Am Acad Dermatol. 1984;11:121-128.
  3. Kumar M, Nair V, Mishra L, et al. Gingival hyperplasia—a clue to the diagnosis of acute leukemia? Arch Oral Sci Res. 2012;2:165-168.
  4. Winfield H, Smoller B. Other lymphoproliferative and myeloproliferative diseases. In: Bolognia JL, Jorizzo JL, Schaffer JV, eds. Dermatology. 3rd ed. Philadelphia, PA: Mosby/Elsevier; 2012:2037-2048.
  5. Babina T, Miller L, Thomas B. Leukemia cutis. J Drugs Dermatol. 2012;11:416-417.
  6. Tziotzios C, Makrygeorgou A. Leukemia cutis. Cleve Clin J Med. 2011;78:226-227.
  7. Ratnam KV, Khor CJL, Su WPD. Leukemia cutis. Dermatol Clin 1994;12:419-431.
  8. Cho-Vega JH, Medeiros LJ, Prieto VG, et al. Leukemia cutis. Am J Clin Pathol. 2008;129:130-142.
  9. Buechner SA, Li CY, Su WP. Leukemia cutis. a histopathologic study of 42 cases. Am J Dermatopathol. 1985;7:109-119.
  10. Wagner G, Fenchel K, Back W, et al. Leukemia cutis—epidemiology, clinical presentation, and differential diagnoses. J Dtsch Dermatol Ges. 2012;10:27-36.
  11. O’Donnell MR, Abboud CN, Altman J, et al. NCCN Clinical Practice Guidelines acute myeloid leukemia. J Natl Compr Canc Netw. 2012;10:984-1021.
  12. Marcucci G, Bloomfield CD. Acute myeloid leukemia. In: Kasper DL, Fauci AS, Hauser SL, et al, eds. Harrison’s Principles of Internal Medicine. 19th ed. New York, NY: McGraw-Hill; 2015:678-686.
  13. Aster JC, DeAngelo DJ. Acute leukemias. In: Bunn HF, Aster JC, eds. Pathophysiology of Blood Disorders. New York, NY: McGraw-Hill; 2010:244-259.
  14. Damon LE, Andreadis C. Blood disorders. In: Papadakis MA, McPhee SJ, Rabow MW, eds. Current Medical Diagnosis & Treatment 2016. New York, NY: McGraw-Hill; 2016:495-541.
  15. Parikh SA, Jabbour E, Koller CA. Adult acute myeloid leukemia. In: Kantarjian HM, Wolff RA, eds. The MD Anderson Manual of Medical Oncology. 2nd ed. New York, NY: McGraw-Hill; 2011:15-32.
References
  1. Rao AG, Danturty I. Leukemia cutis. Indian J Dermatol. 2012;57:504.
  2. Su WPD, Buechner SA, Chin-Yang L. Clinicopathologic correlations in leukemia cutis. J Am Acad Dermatol. 1984;11:121-128.
  3. Kumar M, Nair V, Mishra L, et al. Gingival hyperplasia—a clue to the diagnosis of acute leukemia? Arch Oral Sci Res. 2012;2:165-168.
  4. Winfield H, Smoller B. Other lymphoproliferative and myeloproliferative diseases. In: Bolognia JL, Jorizzo JL, Schaffer JV, eds. Dermatology. 3rd ed. Philadelphia, PA: Mosby/Elsevier; 2012:2037-2048.
  5. Babina T, Miller L, Thomas B. Leukemia cutis. J Drugs Dermatol. 2012;11:416-417.
  6. Tziotzios C, Makrygeorgou A. Leukemia cutis. Cleve Clin J Med. 2011;78:226-227.
  7. Ratnam KV, Khor CJL, Su WPD. Leukemia cutis. Dermatol Clin 1994;12:419-431.
  8. Cho-Vega JH, Medeiros LJ, Prieto VG, et al. Leukemia cutis. Am J Clin Pathol. 2008;129:130-142.
  9. Buechner SA, Li CY, Su WP. Leukemia cutis. a histopathologic study of 42 cases. Am J Dermatopathol. 1985;7:109-119.
  10. Wagner G, Fenchel K, Back W, et al. Leukemia cutis—epidemiology, clinical presentation, and differential diagnoses. J Dtsch Dermatol Ges. 2012;10:27-36.
  11. O’Donnell MR, Abboud CN, Altman J, et al. NCCN Clinical Practice Guidelines acute myeloid leukemia. J Natl Compr Canc Netw. 2012;10:984-1021.
  12. Marcucci G, Bloomfield CD. Acute myeloid leukemia. In: Kasper DL, Fauci AS, Hauser SL, et al, eds. Harrison’s Principles of Internal Medicine. 19th ed. New York, NY: McGraw-Hill; 2015:678-686.
  13. Aster JC, DeAngelo DJ. Acute leukemias. In: Bunn HF, Aster JC, eds. Pathophysiology of Blood Disorders. New York, NY: McGraw-Hill; 2010:244-259.
  14. Damon LE, Andreadis C. Blood disorders. In: Papadakis MA, McPhee SJ, Rabow MW, eds. Current Medical Diagnosis & Treatment 2016. New York, NY: McGraw-Hill; 2016:495-541.
  15. Parikh SA, Jabbour E, Koller CA. Adult acute myeloid leukemia. In: Kantarjian HM, Wolff RA, eds. The MD Anderson Manual of Medical Oncology. 2nd ed. New York, NY: McGraw-Hill; 2011:15-32.
Issue
Cutis - 102(4)
Issue
Cutis - 102(4)
Page Number
266, 271-272
Page Number
266, 271-272
Publications
Publications
Topics
Article Type
Display Headline
Leukemia Cutis in Acute Myeloid Leukemia Signifies a Poor Prognosis
Display Headline
Leukemia Cutis in Acute Myeloid Leukemia Signifies a Poor Prognosis
Sections
Inside the Article

Practice Points

  • Leukemia cutis (LC) describes cutaneous and/or subcutaneous infiltration by leukemic cells and most commonly occurs in patients with acute myeloid leukemia.
  • The vast majority of patients presenting with LC already have systemic involvement.
  • Cutaneous presentation of LC is diverse, thus diagnosis often is dependent on immunohisto-chemical findings.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Article PDF Media

FDA approves cemiplimab for advanced cutaneous squamous cell carcinoma

Article Type
Changed
Fri, 01/18/2019 - 17:59

 

The U.S. Food and Drug Administration has approved cemiplimab-rwlc (Libtayo) for the treatment of metastatic or locally advanced cutaneous squamous cell carcinoma, the agency announced in a press release.

The monoclonal antibody drug – a checkpoint inhibitor that blocks the PD-1 pathway – is the first treatment to be approved specifically for advanced cutaneous squamous cell carcinoma in patients who are not candidates for curative surgery or curative radiation.

The approval was granted based on data from two open-label clinical trials involving a total of 108 patients: the phase 2 EMPOWER-CSCC-1 trial (NCT02760498) and two expansion cohorts from an open-label, nonrandomized phase 1 trial.

These trials, which included 75 patients with metastatic disease and 33 with locally advanced disease, found an overall response rate of 47.2%, and most of those patients still showed ongoing responses at the time of data analysis. Among patients with metastatic disease, 5% had a complete response, according to a press release from the manufacturer, Sanofi.

This is the sixth FDA approval for a checkpoint inhibitor targeting the PD-1/PD-L1 pathway. The drug was evaluated under the FDA’s Priority Review program for drugs that represent significant improvements in the safety or effectiveness of treatments for serious conditions. Manufacturer Sanofi was granted Breakthrough Therapy designation for cemiplimab in 2017 for advanced cutaneous squamous cell carcinoma, and the drug is also being reviewed by the European Medicines Agency.

Cemiplimab is administered as a 350-mg intravenous therapy every 3 weeks – costing $9,100 per treatment – until the disease progresses or patients experience unacceptable toxicity, according to the manufacturer. The most common side effects include fatigue, rash and diarrhea, but more serious adverse events can include immune-mediated reactions such as pneumonitis, colitis, hepatitis, endocrinopathies, and skin and kidney problems.

Publications
Topics
Sections

 

The U.S. Food and Drug Administration has approved cemiplimab-rwlc (Libtayo) for the treatment of metastatic or locally advanced cutaneous squamous cell carcinoma, the agency announced in a press release.

The monoclonal antibody drug – a checkpoint inhibitor that blocks the PD-1 pathway – is the first treatment to be approved specifically for advanced cutaneous squamous cell carcinoma in patients who are not candidates for curative surgery or curative radiation.

The approval was granted based on data from two open-label clinical trials involving a total of 108 patients: the phase 2 EMPOWER-CSCC-1 trial (NCT02760498) and two expansion cohorts from an open-label, nonrandomized phase 1 trial.

These trials, which included 75 patients with metastatic disease and 33 with locally advanced disease, found an overall response rate of 47.2%, and most of those patients still showed ongoing responses at the time of data analysis. Among patients with metastatic disease, 5% had a complete response, according to a press release from the manufacturer, Sanofi.

This is the sixth FDA approval for a checkpoint inhibitor targeting the PD-1/PD-L1 pathway. The drug was evaluated under the FDA’s Priority Review program for drugs that represent significant improvements in the safety or effectiveness of treatments for serious conditions. Manufacturer Sanofi was granted Breakthrough Therapy designation for cemiplimab in 2017 for advanced cutaneous squamous cell carcinoma, and the drug is also being reviewed by the European Medicines Agency.

Cemiplimab is administered as a 350-mg intravenous therapy every 3 weeks – costing $9,100 per treatment – until the disease progresses or patients experience unacceptable toxicity, according to the manufacturer. The most common side effects include fatigue, rash and diarrhea, but more serious adverse events can include immune-mediated reactions such as pneumonitis, colitis, hepatitis, endocrinopathies, and skin and kidney problems.

 

The U.S. Food and Drug Administration has approved cemiplimab-rwlc (Libtayo) for the treatment of metastatic or locally advanced cutaneous squamous cell carcinoma, the agency announced in a press release.

The monoclonal antibody drug – a checkpoint inhibitor that blocks the PD-1 pathway – is the first treatment to be approved specifically for advanced cutaneous squamous cell carcinoma in patients who are not candidates for curative surgery or curative radiation.

The approval was granted based on data from two open-label clinical trials involving a total of 108 patients: the phase 2 EMPOWER-CSCC-1 trial (NCT02760498) and two expansion cohorts from an open-label, nonrandomized phase 1 trial.

These trials, which included 75 patients with metastatic disease and 33 with locally advanced disease, found an overall response rate of 47.2%, and most of those patients still showed ongoing responses at the time of data analysis. Among patients with metastatic disease, 5% had a complete response, according to a press release from the manufacturer, Sanofi.

This is the sixth FDA approval for a checkpoint inhibitor targeting the PD-1/PD-L1 pathway. The drug was evaluated under the FDA’s Priority Review program for drugs that represent significant improvements in the safety or effectiveness of treatments for serious conditions. Manufacturer Sanofi was granted Breakthrough Therapy designation for cemiplimab in 2017 for advanced cutaneous squamous cell carcinoma, and the drug is also being reviewed by the European Medicines Agency.

Cemiplimab is administered as a 350-mg intravenous therapy every 3 weeks – costing $9,100 per treatment – until the disease progresses or patients experience unacceptable toxicity, according to the manufacturer. The most common side effects include fatigue, rash and diarrhea, but more serious adverse events can include immune-mediated reactions such as pneumonitis, colitis, hepatitis, endocrinopathies, and skin and kidney problems.

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

Acral Cutaneous Metastasis From a Primary Breast Carcinoma Following Chemotherapy With Bevacizumab and Paclitaxel

Article Type
Changed
Thu, 01/10/2019 - 13:53
Display Headline
Acral Cutaneous Metastasis From a Primary Breast Carcinoma Following Chemotherapy With Bevacizumab and Paclitaxel

Cutaneous metastasis of internal malignancy is a relatively uncommon phenomenon, with an overall incidence of 5.3% in cancer patients.1 Cutaneous involvement typically occurs late in the course of disease but can occasionally be the first extranodal sign of metastatic disease. Breast cancer has the highest rate of cutaneous metastasis, most often involving the chest wall1; however, cutaneous metastasis to the acral sites is exceedingly rare. The hand is the site of 0.1% of all metastatic lesions, with only 10% of these being cutaneous lesions and the remaining 90% being osseous metastases.2 Herein, we report a case of multiple cutaneous metastases to acral sites involving the palmar and plantar surfaces of the hands and feet.

Case Report

A 54-year-old black woman with a history of stage IV carcinoma of the breast was admitted to the university medical center with exquisitely painful cutaneous nodules on the hands and feet of 5 weeks’ duration that had started to cause difficulty with walking and daily activities. The patient reported that the breast carcinoma had initially been diagnosed in Nigeria 2 years prior, but she did not receive treatment until moving to the United States. She received a total of 4 cycles of chemotherapy with paclitaxel and bevacizumab, which was discontinued 6 weeks prior to admission due to pain in the lower extremities that was thought to be secondary to neuropathy. One week after discontinuation of chemotherapy, the patient reported increasing pain in the extremities and new-onset painful nodules on the hands and feet. Treatment with gabapentin as well as several courses of antibiotics failed to improve the condition.

She was admitted for symptomatic pain control and a dermatology consultation. Physical examination revealed multiple firm, tender, subcutaneous nodules on the volar surfaces of the soles, toes, palms, and fingertips (Figure 1). A nodule also was noted on the scalp. A punch biopsy of a nodule on the right fourth finger revealed a dermal carcinoma (Figure 2). On immunohistochemistry, the tumor stained positive for cytokeratin 5/6, cytokeratin 7, and gross cystic disease fluid protein 15. It did not demonstrate connection to the epidermis or adnexal structures. Although the tumor did not express estrogen or progesterone receptors, the findings were compatible with metastasis from the patient’s primary breast carcinoma with poor differentiation. A biopsy of the primary breast carcinoma was not available for review from Nigeria.

Figure1
Figure 1. Acral cutaneous metastasis with numerous painful subcutaneous nodules on the hands and feet (A–D).

Figure2
Figure 2. A punch biopsy of a nodule on the right fourth finger revealed a poorly differentiated metastatic carcinoma of the breast in the dermis (A and B)(H&E, original magnifications ×4 and ×20).

Comment

The majority of cases reporting acral cutaneous metastasis from internal malignancies are unilateral, involving only one extremity. Several hypotheses have been provided, including spread from localized trauma, which causes disruption of blood vessels and consequent extravasation and localization of tumor cells into the extravascular space.3 The distal extremities are particularly vulnerable to trauma, making this hypothesis plausible.

Considering the overall rarity of metastases to acral sites, it is interesting that our patient developed multiple distal nodules on both the hands and feet. The rapid onset of cutaneous nodules shortly after a course of chemotherapy led the team to consider the physiologic effects of paclitaxel and bevacizumab in the etiology of the acral cutaneous metastases. Karamouzis et al3 described a similar case of multiple cutaneous metastases with a bilateral acral distribution. This case also was associated with chemotherapy in the treatment of breast cancer. The authors proposed hand-foot syndrome, a chemotherapy-related eruption localized to acral skin, as a possible mechanism for hematogenous spread of malignant cells.3 The pathogenesis of hand-foot syndrome is not well understood, but the unique anatomy and physiology of acral skin including temperature gradients, rapidly dividing epidermal cells, absence of hair follicles and sebaceous glands, wide dermal papillae, and exposure to high pressures from carrying body weight and repetitive minor trauma may contribute to the localization of signs and symptoms.3,4 Our case supports a chemotherapy-related etiology of acral cutaneous metastasis of a primary breast cancer; however, our patient did not have apparent signs or symptoms of hand-foot syndrome during the course of treatment. We propose that effects of bevacizumab on acral skin may have contributed to the development of our patient’s metastatic pattern.

Bevacizumab, a monoclonal antibody to vascular endothelial growth factor A, has well-known vascular side effects. Unlike the inhibition of vascular endothelial growth factor A provided by the receptor tyrosine kinase inhibitors sorafenib and sunitinib, bevacizumab typically is not associated with hand-foot syndrome.5 However, several cases have been reported with chemotherapy-associated palmoplantar eruptions that resolved after withholding bevacizumab while continuing other chemotherapeutic agents, suggesting that bevacizumab-induced changes in acral skin contributed to the eruption.6 Specific factors that could contribute to acral metastasis in patients taking bevacizumab are endothelial dysfunction and capillary rarefaction of the acral skin, as well as hemorrhage, decreased wound healing, and changes in vascular permeability.5,7

We present a rare case of acral cutaneous metastasis associated with bevacizumab, one of few reported cases associated with a taxane chemotherapeutic agent.3 More cases need to be identified and reported to establish a causative association, if indeed existent, between acral cutaneous metastasis of breast carcinoma and the use of bevacizumab as well as other chemotherapeutic drugs.

References
  1. Krathen RA, Orengo IF, Rosen T. Cutaneous metastasis: a meta-analysis of data. South Med J. 2003;96:164-167.
  2. Wu CY, Gao HW, Huang WH, et al. Infection-like acral cutaneous metastasis as the presenting sign of an occult breast cancer. Clin Exp Dermatol. 2009;34:409-410.
  3. Karamouzis MV, Ardavanis A, Alexopoulos A, et al. Multiple cutaneous acral metastases in a woman with breast adenocarcinoma treated with pegylated liposomal doxorubicin: incidental or aetiological association? Eur J Cancer Care (Engl). 2005;14:267-271.
  4. Nagore E, Insa A, Sanmartin O. Antineoplastic therapy-induced palmar plantar erythrodysesthesia (‘hand-foot’) syndrome. incidence, recognition and management. Am J Clin Dermatol. 2000;1:225-234.
  5. Wozel G, Sticherling M, Schon MP. Cutaneous side effects of inhibition of VEGF signal transduction. J Dtsch Dermatol Ges. 2010;8:243-249.
  6. Munehiro A, Yoneda K, Nakai K, et al. Bevacizumab-induced hand-foot syndrome: circumscribed type. Br J Dermatol. 2010;162:1411-1413.
  7. Mourad JJ, des Guetz G, Debbabi H, et al. Blood pressure rise following angiogenesis inhibition by bevacizumab. a crucial role for microcirculation. Ann Oncol. 2008;19:927-934.
Article PDF
Author and Disclosure Information

Dr. Armstrong is from the Department of Dermatology, University of California, Los Angeles. Dr. Woody is from the Department of Dermatology, Oregon Health + Sciences University, Portland. Dr. Reichenberg is from the Department of Dermatology, University of Texas, Dell Medical School, Austin. Dr. Gavino is from Tru-Skin Dermatology, Cedar Park, Texas.

The authors report no conflict of interest.

Correspondence: Meghan M. Woody, MD, MPH, OHSU Department of Dermatology, Center for Health & Healing, 3303 SW Bond Ave, Bldg 1, Ste 16, Portland, OR 97239 (woodym@ohsu.edu).

Issue
Cutis - 102(3)
Publications
Topics
Page Number
E12-E14
Sections
Author and Disclosure Information

Dr. Armstrong is from the Department of Dermatology, University of California, Los Angeles. Dr. Woody is from the Department of Dermatology, Oregon Health + Sciences University, Portland. Dr. Reichenberg is from the Department of Dermatology, University of Texas, Dell Medical School, Austin. Dr. Gavino is from Tru-Skin Dermatology, Cedar Park, Texas.

The authors report no conflict of interest.

Correspondence: Meghan M. Woody, MD, MPH, OHSU Department of Dermatology, Center for Health & Healing, 3303 SW Bond Ave, Bldg 1, Ste 16, Portland, OR 97239 (woodym@ohsu.edu).

Author and Disclosure Information

Dr. Armstrong is from the Department of Dermatology, University of California, Los Angeles. Dr. Woody is from the Department of Dermatology, Oregon Health + Sciences University, Portland. Dr. Reichenberg is from the Department of Dermatology, University of Texas, Dell Medical School, Austin. Dr. Gavino is from Tru-Skin Dermatology, Cedar Park, Texas.

The authors report no conflict of interest.

Correspondence: Meghan M. Woody, MD, MPH, OHSU Department of Dermatology, Center for Health & Healing, 3303 SW Bond Ave, Bldg 1, Ste 16, Portland, OR 97239 (woodym@ohsu.edu).

Article PDF
Article PDF

Cutaneous metastasis of internal malignancy is a relatively uncommon phenomenon, with an overall incidence of 5.3% in cancer patients.1 Cutaneous involvement typically occurs late in the course of disease but can occasionally be the first extranodal sign of metastatic disease. Breast cancer has the highest rate of cutaneous metastasis, most often involving the chest wall1; however, cutaneous metastasis to the acral sites is exceedingly rare. The hand is the site of 0.1% of all metastatic lesions, with only 10% of these being cutaneous lesions and the remaining 90% being osseous metastases.2 Herein, we report a case of multiple cutaneous metastases to acral sites involving the palmar and plantar surfaces of the hands and feet.

Case Report

A 54-year-old black woman with a history of stage IV carcinoma of the breast was admitted to the university medical center with exquisitely painful cutaneous nodules on the hands and feet of 5 weeks’ duration that had started to cause difficulty with walking and daily activities. The patient reported that the breast carcinoma had initially been diagnosed in Nigeria 2 years prior, but she did not receive treatment until moving to the United States. She received a total of 4 cycles of chemotherapy with paclitaxel and bevacizumab, which was discontinued 6 weeks prior to admission due to pain in the lower extremities that was thought to be secondary to neuropathy. One week after discontinuation of chemotherapy, the patient reported increasing pain in the extremities and new-onset painful nodules on the hands and feet. Treatment with gabapentin as well as several courses of antibiotics failed to improve the condition.

She was admitted for symptomatic pain control and a dermatology consultation. Physical examination revealed multiple firm, tender, subcutaneous nodules on the volar surfaces of the soles, toes, palms, and fingertips (Figure 1). A nodule also was noted on the scalp. A punch biopsy of a nodule on the right fourth finger revealed a dermal carcinoma (Figure 2). On immunohistochemistry, the tumor stained positive for cytokeratin 5/6, cytokeratin 7, and gross cystic disease fluid protein 15. It did not demonstrate connection to the epidermis or adnexal structures. Although the tumor did not express estrogen or progesterone receptors, the findings were compatible with metastasis from the patient’s primary breast carcinoma with poor differentiation. A biopsy of the primary breast carcinoma was not available for review from Nigeria.

Figure1
Figure 1. Acral cutaneous metastasis with numerous painful subcutaneous nodules on the hands and feet (A–D).

Figure2
Figure 2. A punch biopsy of a nodule on the right fourth finger revealed a poorly differentiated metastatic carcinoma of the breast in the dermis (A and B)(H&E, original magnifications ×4 and ×20).

Comment

The majority of cases reporting acral cutaneous metastasis from internal malignancies are unilateral, involving only one extremity. Several hypotheses have been provided, including spread from localized trauma, which causes disruption of blood vessels and consequent extravasation and localization of tumor cells into the extravascular space.3 The distal extremities are particularly vulnerable to trauma, making this hypothesis plausible.

Considering the overall rarity of metastases to acral sites, it is interesting that our patient developed multiple distal nodules on both the hands and feet. The rapid onset of cutaneous nodules shortly after a course of chemotherapy led the team to consider the physiologic effects of paclitaxel and bevacizumab in the etiology of the acral cutaneous metastases. Karamouzis et al3 described a similar case of multiple cutaneous metastases with a bilateral acral distribution. This case also was associated with chemotherapy in the treatment of breast cancer. The authors proposed hand-foot syndrome, a chemotherapy-related eruption localized to acral skin, as a possible mechanism for hematogenous spread of malignant cells.3 The pathogenesis of hand-foot syndrome is not well understood, but the unique anatomy and physiology of acral skin including temperature gradients, rapidly dividing epidermal cells, absence of hair follicles and sebaceous glands, wide dermal papillae, and exposure to high pressures from carrying body weight and repetitive minor trauma may contribute to the localization of signs and symptoms.3,4 Our case supports a chemotherapy-related etiology of acral cutaneous metastasis of a primary breast cancer; however, our patient did not have apparent signs or symptoms of hand-foot syndrome during the course of treatment. We propose that effects of bevacizumab on acral skin may have contributed to the development of our patient’s metastatic pattern.

Bevacizumab, a monoclonal antibody to vascular endothelial growth factor A, has well-known vascular side effects. Unlike the inhibition of vascular endothelial growth factor A provided by the receptor tyrosine kinase inhibitors sorafenib and sunitinib, bevacizumab typically is not associated with hand-foot syndrome.5 However, several cases have been reported with chemotherapy-associated palmoplantar eruptions that resolved after withholding bevacizumab while continuing other chemotherapeutic agents, suggesting that bevacizumab-induced changes in acral skin contributed to the eruption.6 Specific factors that could contribute to acral metastasis in patients taking bevacizumab are endothelial dysfunction and capillary rarefaction of the acral skin, as well as hemorrhage, decreased wound healing, and changes in vascular permeability.5,7

We present a rare case of acral cutaneous metastasis associated with bevacizumab, one of few reported cases associated with a taxane chemotherapeutic agent.3 More cases need to be identified and reported to establish a causative association, if indeed existent, between acral cutaneous metastasis of breast carcinoma and the use of bevacizumab as well as other chemotherapeutic drugs.

Cutaneous metastasis of internal malignancy is a relatively uncommon phenomenon, with an overall incidence of 5.3% in cancer patients.1 Cutaneous involvement typically occurs late in the course of disease but can occasionally be the first extranodal sign of metastatic disease. Breast cancer has the highest rate of cutaneous metastasis, most often involving the chest wall1; however, cutaneous metastasis to the acral sites is exceedingly rare. The hand is the site of 0.1% of all metastatic lesions, with only 10% of these being cutaneous lesions and the remaining 90% being osseous metastases.2 Herein, we report a case of multiple cutaneous metastases to acral sites involving the palmar and plantar surfaces of the hands and feet.

Case Report

A 54-year-old black woman with a history of stage IV carcinoma of the breast was admitted to the university medical center with exquisitely painful cutaneous nodules on the hands and feet of 5 weeks’ duration that had started to cause difficulty with walking and daily activities. The patient reported that the breast carcinoma had initially been diagnosed in Nigeria 2 years prior, but she did not receive treatment until moving to the United States. She received a total of 4 cycles of chemotherapy with paclitaxel and bevacizumab, which was discontinued 6 weeks prior to admission due to pain in the lower extremities that was thought to be secondary to neuropathy. One week after discontinuation of chemotherapy, the patient reported increasing pain in the extremities and new-onset painful nodules on the hands and feet. Treatment with gabapentin as well as several courses of antibiotics failed to improve the condition.

She was admitted for symptomatic pain control and a dermatology consultation. Physical examination revealed multiple firm, tender, subcutaneous nodules on the volar surfaces of the soles, toes, palms, and fingertips (Figure 1). A nodule also was noted on the scalp. A punch biopsy of a nodule on the right fourth finger revealed a dermal carcinoma (Figure 2). On immunohistochemistry, the tumor stained positive for cytokeratin 5/6, cytokeratin 7, and gross cystic disease fluid protein 15. It did not demonstrate connection to the epidermis or adnexal structures. Although the tumor did not express estrogen or progesterone receptors, the findings were compatible with metastasis from the patient’s primary breast carcinoma with poor differentiation. A biopsy of the primary breast carcinoma was not available for review from Nigeria.

Figure1
Figure 1. Acral cutaneous metastasis with numerous painful subcutaneous nodules on the hands and feet (A–D).

Figure2
Figure 2. A punch biopsy of a nodule on the right fourth finger revealed a poorly differentiated metastatic carcinoma of the breast in the dermis (A and B)(H&E, original magnifications ×4 and ×20).

Comment

The majority of cases reporting acral cutaneous metastasis from internal malignancies are unilateral, involving only one extremity. Several hypotheses have been provided, including spread from localized trauma, which causes disruption of blood vessels and consequent extravasation and localization of tumor cells into the extravascular space.3 The distal extremities are particularly vulnerable to trauma, making this hypothesis plausible.

Considering the overall rarity of metastases to acral sites, it is interesting that our patient developed multiple distal nodules on both the hands and feet. The rapid onset of cutaneous nodules shortly after a course of chemotherapy led the team to consider the physiologic effects of paclitaxel and bevacizumab in the etiology of the acral cutaneous metastases. Karamouzis et al3 described a similar case of multiple cutaneous metastases with a bilateral acral distribution. This case also was associated with chemotherapy in the treatment of breast cancer. The authors proposed hand-foot syndrome, a chemotherapy-related eruption localized to acral skin, as a possible mechanism for hematogenous spread of malignant cells.3 The pathogenesis of hand-foot syndrome is not well understood, but the unique anatomy and physiology of acral skin including temperature gradients, rapidly dividing epidermal cells, absence of hair follicles and sebaceous glands, wide dermal papillae, and exposure to high pressures from carrying body weight and repetitive minor trauma may contribute to the localization of signs and symptoms.3,4 Our case supports a chemotherapy-related etiology of acral cutaneous metastasis of a primary breast cancer; however, our patient did not have apparent signs or symptoms of hand-foot syndrome during the course of treatment. We propose that effects of bevacizumab on acral skin may have contributed to the development of our patient’s metastatic pattern.

Bevacizumab, a monoclonal antibody to vascular endothelial growth factor A, has well-known vascular side effects. Unlike the inhibition of vascular endothelial growth factor A provided by the receptor tyrosine kinase inhibitors sorafenib and sunitinib, bevacizumab typically is not associated with hand-foot syndrome.5 However, several cases have been reported with chemotherapy-associated palmoplantar eruptions that resolved after withholding bevacizumab while continuing other chemotherapeutic agents, suggesting that bevacizumab-induced changes in acral skin contributed to the eruption.6 Specific factors that could contribute to acral metastasis in patients taking bevacizumab are endothelial dysfunction and capillary rarefaction of the acral skin, as well as hemorrhage, decreased wound healing, and changes in vascular permeability.5,7

We present a rare case of acral cutaneous metastasis associated with bevacizumab, one of few reported cases associated with a taxane chemotherapeutic agent.3 More cases need to be identified and reported to establish a causative association, if indeed existent, between acral cutaneous metastasis of breast carcinoma and the use of bevacizumab as well as other chemotherapeutic drugs.

References
  1. Krathen RA, Orengo IF, Rosen T. Cutaneous metastasis: a meta-analysis of data. South Med J. 2003;96:164-167.
  2. Wu CY, Gao HW, Huang WH, et al. Infection-like acral cutaneous metastasis as the presenting sign of an occult breast cancer. Clin Exp Dermatol. 2009;34:409-410.
  3. Karamouzis MV, Ardavanis A, Alexopoulos A, et al. Multiple cutaneous acral metastases in a woman with breast adenocarcinoma treated with pegylated liposomal doxorubicin: incidental or aetiological association? Eur J Cancer Care (Engl). 2005;14:267-271.
  4. Nagore E, Insa A, Sanmartin O. Antineoplastic therapy-induced palmar plantar erythrodysesthesia (‘hand-foot’) syndrome. incidence, recognition and management. Am J Clin Dermatol. 2000;1:225-234.
  5. Wozel G, Sticherling M, Schon MP. Cutaneous side effects of inhibition of VEGF signal transduction. J Dtsch Dermatol Ges. 2010;8:243-249.
  6. Munehiro A, Yoneda K, Nakai K, et al. Bevacizumab-induced hand-foot syndrome: circumscribed type. Br J Dermatol. 2010;162:1411-1413.
  7. Mourad JJ, des Guetz G, Debbabi H, et al. Blood pressure rise following angiogenesis inhibition by bevacizumab. a crucial role for microcirculation. Ann Oncol. 2008;19:927-934.
References
  1. Krathen RA, Orengo IF, Rosen T. Cutaneous metastasis: a meta-analysis of data. South Med J. 2003;96:164-167.
  2. Wu CY, Gao HW, Huang WH, et al. Infection-like acral cutaneous metastasis as the presenting sign of an occult breast cancer. Clin Exp Dermatol. 2009;34:409-410.
  3. Karamouzis MV, Ardavanis A, Alexopoulos A, et al. Multiple cutaneous acral metastases in a woman with breast adenocarcinoma treated with pegylated liposomal doxorubicin: incidental or aetiological association? Eur J Cancer Care (Engl). 2005;14:267-271.
  4. Nagore E, Insa A, Sanmartin O. Antineoplastic therapy-induced palmar plantar erythrodysesthesia (‘hand-foot’) syndrome. incidence, recognition and management. Am J Clin Dermatol. 2000;1:225-234.
  5. Wozel G, Sticherling M, Schon MP. Cutaneous side effects of inhibition of VEGF signal transduction. J Dtsch Dermatol Ges. 2010;8:243-249.
  6. Munehiro A, Yoneda K, Nakai K, et al. Bevacizumab-induced hand-foot syndrome: circumscribed type. Br J Dermatol. 2010;162:1411-1413.
  7. Mourad JJ, des Guetz G, Debbabi H, et al. Blood pressure rise following angiogenesis inhibition by bevacizumab. a crucial role for microcirculation. Ann Oncol. 2008;19:927-934.
Issue
Cutis - 102(3)
Issue
Cutis - 102(3)
Page Number
E12-E14
Page Number
E12-E14
Publications
Publications
Topics
Article Type
Display Headline
Acral Cutaneous Metastasis From a Primary Breast Carcinoma Following Chemotherapy With Bevacizumab and Paclitaxel
Display Headline
Acral Cutaneous Metastasis From a Primary Breast Carcinoma Following Chemotherapy With Bevacizumab and Paclitaxel
Sections
Inside the Article

Practice Points

  • Cutaneous involvement of internal malignancy typically occurs late in the disease course but can occasionally be the first extranodal sign of metastatic disease.
  • Acral cutaneous metastasis from internal malignancies typically is unilateral, involving only one extremity; however, this case demonstrates involvement on both the hands and feet.
  • This case support a chemotherapy-related etiology of acral cutaneous metastasis of a primary breast cancer.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Article PDF Media

Skin signs may be good omens during cancer therapy

Article Type
Changed
Mon, 01/14/2019 - 10:31

 

Signs of efficacy of anti-cancer therapies may be only skin deep, results of a retrospective review indicate.

Cutaneous toxicities such as vitiligo, rash, alopecia, and nail toxicities may be early signs of efficacy of targeted therapies, immunotherapy, or cytotoxic chemotherapy, according to Alexandra K. Rzepecki, of the University of Michigan, and her coauthors from Albert Einstein Medical College in the Bronx, New York.

“Because cutaneous toxicities are a clinically visible parameter, they may alert clinicians to the possibility of treatment success or failure in a rapid, cost-effective, and noninvasive manner,” they wrote. The report is in the Journal of the American Academy of Dermatology.

The investigators reviewed the medical literature for clinical studies of three major classes of anti-cancer therapies that included data on associations between cutaneous toxicities and clinical outcomes such progression-free survival (PFS) overall survival (OS).

The drug classes and their associations with cutaneous toxicities and clinical outcomes were as follows:

  • Targeted therapies, including tyrosine kinase inhibitors targeting the epidermal growth factor receptor (EGFR) such as cetuximab (Erbitux) and erlotinib (Tarceva), and multikinase targeted agents such as sorafenib (Nexavar) and sunitinib (Sutent). Toxicities associated with clinical benefit from EGFR inhibitors include rash, xerosis, leukocytoclastic vasculitis, paronychia, and pruritus, whereas skin toxicities associated with the multikinase inhibitors trended toward the hand-foot syndrome and hand-foot skin reaction.
  • Immunotherapies included blockers of cytotoxic T-lymphocyte associated protein 4 (CTLA4) such as ipilimumab (Yervoy) and inhibitors of programmed death 1 protein (PD-1) and its ligand 1 (PD-L1) such as nivolumab (Opdivo), pembrolizumab (Keytruda), and atezolizumab (Tecentriq). In studies of pembrolizumab for various malignancies, rash or vitiligo was an independent prognostic factor for longer OS, a higher proportion of objective responses, and longer PFS. Similar associations were seen with nivolumab, with the additional association of hair repigmentation among patients with non–small-cell lung cancer being associated with stable disease responses or better. Among patients with melanoma treated with ipilimumab, hair depigmentation correlated with durable responses.
  • Cytotoxic chemotherapy agents included the anthracycline doxorubicin, taxanes such as paclitaxel and docetaxel, platinum agents (cisplatin and carboplatin), and fluoropyrimidines such as capecitabine. Patients treated for various cancers with doxorubicin who had alopecia were significantly more likely to have clinical remissions than were patients who did not lose their hair, and patients treated with this agent who developed hand-foot syndrome had significantly longer PFS. For patients treated with docetaxel, severity of nail changes and/or development of nail alterations were associated with both improved OS and PFS. Patients treated with the combination of paclitaxel and a platinum agent who developed grade 2 or greater alopecia up to cycle 3 had significantly longer OS than did patients who had hair loss later in the course of therapy. Patients treated with capecitabine who developed had hand-foot skin reactions had improved progression-free and disease-free survival.

“Although further studies are needed to better evaluate these promising associations, vigilant monitoring of cutaneous toxicities should be a priority, as their development may indicate a favorable response to treatment. Dermatologists have a unique opportunity to collaborate with oncologists to help identify and manage these toxicities, thereby allowing patients to receive life-prolonging anticancer therapy while minimizing dose reduction or interruption of their treatment,” the authors wrote.

They reported no study funding source and no conflicts of interest.

SOURCE: Rzepecki A, et al. J Am Acad Dermatol. 2018;79:545-555.

Publications
Topics
Sections

 

Signs of efficacy of anti-cancer therapies may be only skin deep, results of a retrospective review indicate.

Cutaneous toxicities such as vitiligo, rash, alopecia, and nail toxicities may be early signs of efficacy of targeted therapies, immunotherapy, or cytotoxic chemotherapy, according to Alexandra K. Rzepecki, of the University of Michigan, and her coauthors from Albert Einstein Medical College in the Bronx, New York.

“Because cutaneous toxicities are a clinically visible parameter, they may alert clinicians to the possibility of treatment success or failure in a rapid, cost-effective, and noninvasive manner,” they wrote. The report is in the Journal of the American Academy of Dermatology.

The investigators reviewed the medical literature for clinical studies of three major classes of anti-cancer therapies that included data on associations between cutaneous toxicities and clinical outcomes such progression-free survival (PFS) overall survival (OS).

The drug classes and their associations with cutaneous toxicities and clinical outcomes were as follows:

  • Targeted therapies, including tyrosine kinase inhibitors targeting the epidermal growth factor receptor (EGFR) such as cetuximab (Erbitux) and erlotinib (Tarceva), and multikinase targeted agents such as sorafenib (Nexavar) and sunitinib (Sutent). Toxicities associated with clinical benefit from EGFR inhibitors include rash, xerosis, leukocytoclastic vasculitis, paronychia, and pruritus, whereas skin toxicities associated with the multikinase inhibitors trended toward the hand-foot syndrome and hand-foot skin reaction.
  • Immunotherapies included blockers of cytotoxic T-lymphocyte associated protein 4 (CTLA4) such as ipilimumab (Yervoy) and inhibitors of programmed death 1 protein (PD-1) and its ligand 1 (PD-L1) such as nivolumab (Opdivo), pembrolizumab (Keytruda), and atezolizumab (Tecentriq). In studies of pembrolizumab for various malignancies, rash or vitiligo was an independent prognostic factor for longer OS, a higher proportion of objective responses, and longer PFS. Similar associations were seen with nivolumab, with the additional association of hair repigmentation among patients with non–small-cell lung cancer being associated with stable disease responses or better. Among patients with melanoma treated with ipilimumab, hair depigmentation correlated with durable responses.
  • Cytotoxic chemotherapy agents included the anthracycline doxorubicin, taxanes such as paclitaxel and docetaxel, platinum agents (cisplatin and carboplatin), and fluoropyrimidines such as capecitabine. Patients treated for various cancers with doxorubicin who had alopecia were significantly more likely to have clinical remissions than were patients who did not lose their hair, and patients treated with this agent who developed hand-foot syndrome had significantly longer PFS. For patients treated with docetaxel, severity of nail changes and/or development of nail alterations were associated with both improved OS and PFS. Patients treated with the combination of paclitaxel and a platinum agent who developed grade 2 or greater alopecia up to cycle 3 had significantly longer OS than did patients who had hair loss later in the course of therapy. Patients treated with capecitabine who developed had hand-foot skin reactions had improved progression-free and disease-free survival.

“Although further studies are needed to better evaluate these promising associations, vigilant monitoring of cutaneous toxicities should be a priority, as their development may indicate a favorable response to treatment. Dermatologists have a unique opportunity to collaborate with oncologists to help identify and manage these toxicities, thereby allowing patients to receive life-prolonging anticancer therapy while minimizing dose reduction or interruption of their treatment,” the authors wrote.

They reported no study funding source and no conflicts of interest.

SOURCE: Rzepecki A, et al. J Am Acad Dermatol. 2018;79:545-555.

 

Signs of efficacy of anti-cancer therapies may be only skin deep, results of a retrospective review indicate.

Cutaneous toxicities such as vitiligo, rash, alopecia, and nail toxicities may be early signs of efficacy of targeted therapies, immunotherapy, or cytotoxic chemotherapy, according to Alexandra K. Rzepecki, of the University of Michigan, and her coauthors from Albert Einstein Medical College in the Bronx, New York.

“Because cutaneous toxicities are a clinically visible parameter, they may alert clinicians to the possibility of treatment success or failure in a rapid, cost-effective, and noninvasive manner,” they wrote. The report is in the Journal of the American Academy of Dermatology.

The investigators reviewed the medical literature for clinical studies of three major classes of anti-cancer therapies that included data on associations between cutaneous toxicities and clinical outcomes such progression-free survival (PFS) overall survival (OS).

The drug classes and their associations with cutaneous toxicities and clinical outcomes were as follows:

  • Targeted therapies, including tyrosine kinase inhibitors targeting the epidermal growth factor receptor (EGFR) such as cetuximab (Erbitux) and erlotinib (Tarceva), and multikinase targeted agents such as sorafenib (Nexavar) and sunitinib (Sutent). Toxicities associated with clinical benefit from EGFR inhibitors include rash, xerosis, leukocytoclastic vasculitis, paronychia, and pruritus, whereas skin toxicities associated with the multikinase inhibitors trended toward the hand-foot syndrome and hand-foot skin reaction.
  • Immunotherapies included blockers of cytotoxic T-lymphocyte associated protein 4 (CTLA4) such as ipilimumab (Yervoy) and inhibitors of programmed death 1 protein (PD-1) and its ligand 1 (PD-L1) such as nivolumab (Opdivo), pembrolizumab (Keytruda), and atezolizumab (Tecentriq). In studies of pembrolizumab for various malignancies, rash or vitiligo was an independent prognostic factor for longer OS, a higher proportion of objective responses, and longer PFS. Similar associations were seen with nivolumab, with the additional association of hair repigmentation among patients with non–small-cell lung cancer being associated with stable disease responses or better. Among patients with melanoma treated with ipilimumab, hair depigmentation correlated with durable responses.
  • Cytotoxic chemotherapy agents included the anthracycline doxorubicin, taxanes such as paclitaxel and docetaxel, platinum agents (cisplatin and carboplatin), and fluoropyrimidines such as capecitabine. Patients treated for various cancers with doxorubicin who had alopecia were significantly more likely to have clinical remissions than were patients who did not lose their hair, and patients treated with this agent who developed hand-foot syndrome had significantly longer PFS. For patients treated with docetaxel, severity of nail changes and/or development of nail alterations were associated with both improved OS and PFS. Patients treated with the combination of paclitaxel and a platinum agent who developed grade 2 or greater alopecia up to cycle 3 had significantly longer OS than did patients who had hair loss later in the course of therapy. Patients treated with capecitabine who developed had hand-foot skin reactions had improved progression-free and disease-free survival.

“Although further studies are needed to better evaluate these promising associations, vigilant monitoring of cutaneous toxicities should be a priority, as their development may indicate a favorable response to treatment. Dermatologists have a unique opportunity to collaborate with oncologists to help identify and manage these toxicities, thereby allowing patients to receive life-prolonging anticancer therapy while minimizing dose reduction or interruption of their treatment,” the authors wrote.

They reported no study funding source and no conflicts of interest.

SOURCE: Rzepecki A, et al. J Am Acad Dermatol. 2018;79:545-555.

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

FROM JOURNAL OF THE AMERICAN ACADEMY OF DERMATOLOGY

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

 

Key clinical point: Cutaneous adverse events may be early signs of drug efficacy in patients treated for various cancers.

Major finding: Cutaneous toxicities with targeted therapies, immunotherapy, and cytotoxic drugs were associated in multiple studies with improved outcomes, including progression-free and overall survival.

Study details: Retrospective review of medical literature for clinical studies reporting associations between cutaneous toxicities and clinical outcomes of cancer therapy.

Disclosures: The authors reported no study funding source and no conflicts of interest.

Source: Rzepecki A et al. J Am Acad Dermatol. 2018 Sep;79[3]:545-55.

Disqus Comments
Default
Use ProPublica

Reflectance confocal microscopy: The future looks bright

Article Type
Changed
Mon, 01/14/2019 - 10:31

 

– The future looks bright for reflectance confocal microscopy (RCM) as a tie breaker to decide whether skin lesions that are equivocal on dermoscopy warrant biopsy to rule out malignancy, Ann M. John, MD, asserted at the annual meeting of the American College of Mohs Surgery.

Dr. Ann M. John

“With the advent of dermoscopy, dermatologists were able to elucidate both benign and malignant patterns to help further guide their decision to biopsy or not. This increased diagnostic accuracy of suspicious lesions by 30%, while reducing the benign to malignant ratio of biopsies performed from 18:1 to 4:1. However, there are still lesions that are equivocal on dermoscopy, as we all know, and for this, there’s reflectance confocal microscopy,” observed Dr. John, of Robert Wood Johnson Medical School, New Brunswick, N.J.

RCM is a device technology that’s been cleared by the Food and Drug Administration since 2008 for the imaging of clinically suspicious lesions. It employs laser scanning to assess the light-scattering properties of cells in the epidermis and dermis, generating images with resolution comparable to histology.

RCM took a back seat initially while American dermatologists were gradually coming to embrace dermoscopy, which their European colleagues had done years earlier. Now, with the availability of handheld RCM for use in the dermatology clinic, expect RCM to assume a growing role in daily practice.


To illustrate the power of RCM as a diagnostic aid, she presented a single-center retrospective study of 1,189 clinically suspicious skin lesions that were equivocal on dermoscopy and then assessed using RCM with 1 year of subsequent patient follow-up. Overall, 155 lesions were deemed positive for cancer or atypia by RCM, while 1,034 were determined to be benign. Of those 155, 46 lesions were considered false positives because of their benign appearance on histologic inspection of the biopsy sample. Only 2 of the 1,034 lesions identified as negative by RCM proved to be false negatives on the basis of clinical changes within 1 year.

The overall sensitivity and specificity of RCM was 98.2% and 99.8%, respectively, with a positive predictive value of 70.3% and a negative predictive value of 99.8%.

The entire RCM procedure takes a skilled technician 15-20 minutes per lesion. As a practical matter, other investigators have estimated that RCM results in a cost savings of about $308,000 per million health plan members per year by reducing the need for biopsies (Dermatol Clin. 2016 Oct;34[4]:367-75).

In addition to evaluating clinically suspicious lesions, other situations in which RCM offers practical value include its use directly before the first cut during Mohs surgery in order to determine the margins of atypia; ex vivo imaging of Mohs margins, which has been shown to be comparable with frozen sections in accuracy but takes only one-third of the time; and imaging of biopsied lesions in order to determine the diagnosis relatively quickly, Dr. John noted.

She reported having no financial conflicts regarding her study.

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

 

– The future looks bright for reflectance confocal microscopy (RCM) as a tie breaker to decide whether skin lesions that are equivocal on dermoscopy warrant biopsy to rule out malignancy, Ann M. John, MD, asserted at the annual meeting of the American College of Mohs Surgery.

Dr. Ann M. John

“With the advent of dermoscopy, dermatologists were able to elucidate both benign and malignant patterns to help further guide their decision to biopsy or not. This increased diagnostic accuracy of suspicious lesions by 30%, while reducing the benign to malignant ratio of biopsies performed from 18:1 to 4:1. However, there are still lesions that are equivocal on dermoscopy, as we all know, and for this, there’s reflectance confocal microscopy,” observed Dr. John, of Robert Wood Johnson Medical School, New Brunswick, N.J.

RCM is a device technology that’s been cleared by the Food and Drug Administration since 2008 for the imaging of clinically suspicious lesions. It employs laser scanning to assess the light-scattering properties of cells in the epidermis and dermis, generating images with resolution comparable to histology.

RCM took a back seat initially while American dermatologists were gradually coming to embrace dermoscopy, which their European colleagues had done years earlier. Now, with the availability of handheld RCM for use in the dermatology clinic, expect RCM to assume a growing role in daily practice.


To illustrate the power of RCM as a diagnostic aid, she presented a single-center retrospective study of 1,189 clinically suspicious skin lesions that were equivocal on dermoscopy and then assessed using RCM with 1 year of subsequent patient follow-up. Overall, 155 lesions were deemed positive for cancer or atypia by RCM, while 1,034 were determined to be benign. Of those 155, 46 lesions were considered false positives because of their benign appearance on histologic inspection of the biopsy sample. Only 2 of the 1,034 lesions identified as negative by RCM proved to be false negatives on the basis of clinical changes within 1 year.

The overall sensitivity and specificity of RCM was 98.2% and 99.8%, respectively, with a positive predictive value of 70.3% and a negative predictive value of 99.8%.

The entire RCM procedure takes a skilled technician 15-20 minutes per lesion. As a practical matter, other investigators have estimated that RCM results in a cost savings of about $308,000 per million health plan members per year by reducing the need for biopsies (Dermatol Clin. 2016 Oct;34[4]:367-75).

In addition to evaluating clinically suspicious lesions, other situations in which RCM offers practical value include its use directly before the first cut during Mohs surgery in order to determine the margins of atypia; ex vivo imaging of Mohs margins, which has been shown to be comparable with frozen sections in accuracy but takes only one-third of the time; and imaging of biopsied lesions in order to determine the diagnosis relatively quickly, Dr. John noted.

She reported having no financial conflicts regarding her study.

 

– The future looks bright for reflectance confocal microscopy (RCM) as a tie breaker to decide whether skin lesions that are equivocal on dermoscopy warrant biopsy to rule out malignancy, Ann M. John, MD, asserted at the annual meeting of the American College of Mohs Surgery.

Dr. Ann M. John

“With the advent of dermoscopy, dermatologists were able to elucidate both benign and malignant patterns to help further guide their decision to biopsy or not. This increased diagnostic accuracy of suspicious lesions by 30%, while reducing the benign to malignant ratio of biopsies performed from 18:1 to 4:1. However, there are still lesions that are equivocal on dermoscopy, as we all know, and for this, there’s reflectance confocal microscopy,” observed Dr. John, of Robert Wood Johnson Medical School, New Brunswick, N.J.

RCM is a device technology that’s been cleared by the Food and Drug Administration since 2008 for the imaging of clinically suspicious lesions. It employs laser scanning to assess the light-scattering properties of cells in the epidermis and dermis, generating images with resolution comparable to histology.

RCM took a back seat initially while American dermatologists were gradually coming to embrace dermoscopy, which their European colleagues had done years earlier. Now, with the availability of handheld RCM for use in the dermatology clinic, expect RCM to assume a growing role in daily practice.


To illustrate the power of RCM as a diagnostic aid, she presented a single-center retrospective study of 1,189 clinically suspicious skin lesions that were equivocal on dermoscopy and then assessed using RCM with 1 year of subsequent patient follow-up. Overall, 155 lesions were deemed positive for cancer or atypia by RCM, while 1,034 were determined to be benign. Of those 155, 46 lesions were considered false positives because of their benign appearance on histologic inspection of the biopsy sample. Only 2 of the 1,034 lesions identified as negative by RCM proved to be false negatives on the basis of clinical changes within 1 year.

The overall sensitivity and specificity of RCM was 98.2% and 99.8%, respectively, with a positive predictive value of 70.3% and a negative predictive value of 99.8%.

The entire RCM procedure takes a skilled technician 15-20 minutes per lesion. As a practical matter, other investigators have estimated that RCM results in a cost savings of about $308,000 per million health plan members per year by reducing the need for biopsies (Dermatol Clin. 2016 Oct;34[4]:367-75).

In addition to evaluating clinically suspicious lesions, other situations in which RCM offers practical value include its use directly before the first cut during Mohs surgery in order to determine the margins of atypia; ex vivo imaging of Mohs margins, which has been shown to be comparable with frozen sections in accuracy but takes only one-third of the time; and imaging of biopsied lesions in order to determine the diagnosis relatively quickly, Dr. John noted.

She reported having no financial conflicts regarding her study.

Publications
Publications
Topics
Article Type
Sections
Article Source

REPORTING FROM THE ACMS ANNUAL MEETING

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

 

Key clinical point: The future looks bright for reflectance confocal microscopy in dermatology.

Major finding: The sensitivity and specificity of reflectance confocal microscopy for diagnosis of skin cancer in patients with equivocal dermoscopic findings was 98.2% and 99.8%, respectively.

Study details: This retrospective single center study included 1,189 clinically suspicious skin lesions with equivocal dermoscopy findings, which were then evaluated using reflectance confocal microscopy.

Disclosures: The presenter reported having no financial conflicts regarding her study.

Disqus Comments
Default
Use ProPublica

A Rare Case of Primary Cutaneous Diffuse Large B-Cell Lymphoma, Leg Type

Article Type
Changed
Thu, 01/10/2019 - 13:53

CASE REPORT

A 74-year-old woman presented with a painful lesion on the left lower leg that was getting larger and more edematous and erythematous over the last 5 months. She experienced numbness and burning of the left lower leg 1 year prior to the development of the lesion. A review of her medical history revealed an otherwise healthy woman with no constitutional symptoms of fever, chills, nausea, vomiting, diarrhea, or chest pain. The patient did not exhibit mucosal, genital, or nail involvement. Physical examination revealed a group of four 1-cm, ill-defined, irregularly bordered, violaceous plaques on the left anterior tibial leg with faint surrounding erythematous to violaceous patches (Figure 1). The plaques were tender to palpation with no bleeding or drainage.

Figure1
Figure 1. Primary cutaneous diffuse large B-cell lymphoma, leg type presenting as a violaceous, ill-defined, tender plaque with both surrounding and satellite faint erythematous to violaceous patches.

An 8.0-mm punch biopsy of the lesion was obtained. Hematoxylin and eosin staining on low-power magnification demonstrated a diffuse lymphocytic inflammatory infiltrate in the dermis and subcutis. Notable sparing of the subepidermal area (free grenz zone) was present (Figure 2A). On higher power, centroblasts and immunoblasts were visualized alongside extravasated red blood cells (Figure 2B). A diagnosis of primary cutaneous diffuse large B-cell lymphoma, leg type (DLBCLLT) was made. Various immunohistochemical stains confirmed the diagnosis, including B-cell lymphoma 2 (BCL-2)(Figure 3A) and multiple myeloma oncogene 1 (MUM-1)(Figure 3B), which were highly positive in our patient. The patient had a negative bone marrow biopsy and positron emission tomography scan. She was started on rituximab infusions and multiple radiation treatments. At 2-year follow-up the lymphoma continued to recur despite radiation therapy.

Figure2
Figure 2. Histopathology of primary cutaneous diffuse large B-cell lymphoma, leg type demonstrated a diffuse inflammatory infiltrate in the dermis and subcutis with a subepidermal free grenz zone on low power (A)(H&E). High-power view demonstrated centroblasts and immunoblasts alongside extravasated red blood cells (B)(H&E).

Figure 3. Positive B-cell lymphoma 2 (BCL-2)(A) and multiple myeloma oncogene 1 (MUM-1)(B) immunohistochemical staining confirmed the diagnosis.

COMMENT

Incidence and Clinical Characteristics

Primary cutaneous DLBCLLT is an intermediately aggressive form of primary cutaneous B-cell lymphoma (CBCL) that accounts for approximately 10% to 20% of all primary CBCLs and 1% to 3% of all cutaneous lymphomas.1 Diffuse large B-cell lymphoma, leg type primarily affects elderly patients (median age, 70 years). Women are more commonly affected. Clinically, primary cutaneous DLBCLLT presents as red-brown to bluish nodules or tumors on one or both distal legs. Although referred to as leg-type diffuse large B-cell lymphoma, 10% to 15% of patients have lesions in anatomic areas other than the legs, most commonly the trunk.

Histopathology

The diagnosis of DLBCLLT is best made histologically. There is a dense inflammatory infiltrate present in the dermis and subcutis that may extend upward into the dermoepidermal junction. Often a subepidermal free grenz zone may be seen, and adnexal structures may be destroyed. This infiltrate is composed of confluent sheets of large round cells including centroblasts and immunoblasts.2 Centroblasts are large cells that have nuclei with several small nucleoli adhering to the membrane, while immunoblasts are large round cells containing nuclei with large central nucleoli. Both centroblasts and immunoblasts stain positively for BCL-2. Centrocytes typically are absent. Staining for BCL-2 can be important in distinguishing DLBCLLT from other forms of CBCL. Diffuse large B-cell lymphoma, leg type also can demonstrate clusters of large atypical cells in the epidermis simulating epidermotropism and Pautrier microabscesses. Neoplastic cells in this condition may express monoclonal surface and cytoplasmic immunoglobulins. Primary cutaneous DLBCLLT typically is positive for B-cell markers CD20 and CD79a. Additionally, MUM-1/IRF4 (interferon regulatory factor 4) and forkhead box protein 1 (FOXP1) are strongly expressed by most patients, which helps distinguish it from other forms of CBCL.

Treatment

Diffuse large B-cell lymphoma, leg type is a relatively aggressive form of CBCL that requires more aggressive treatment than the conservative watchful waiting of some of the more indolent forms of primary CBCL. One regimen involves using cyclophosphamide, doxorubicin, vincristine, and prednisone plus rituximab. Local chemotherapy or radiation with rituximab is another treatment option.1,2 In patients with severe comorbidities, rituximab alone may be administered. The prognosis for DLBCLLT is not as favorable as other types of primary CBCL, with an estimated 5-year survival rate of approximately 50%.2

 

 

Differential Diagnosis

Lymphomas are malignancies of the lymphocytes that may be subdivided depending on the organ of origin. Both primary nodal lymphomas and primary cutaneous lymphomas exist. Primary nodal lymphomas arise from the lymph nodes and are divided into Hodgkin and non-Hodgkin lymphomas. There are 2 major types of primary cutaneous lymphomas: cutaneous T-cell lymphoma (CTCL) and CBCL. Most primary cutaneous lymphomas are CTCLs, accounting for 75% to 80%.3

Pseudolymphoma
Pseudolymphoma is an inflammatory condition that may histologically mimic cutaneous lymphoma but has a benign clinical course. Pseudolymphoma is not a specific disease but rather is a reactive lymphoproliferative response to a known or unknown stimulus.4 Pseudolymphoma can be broken down into 2 or 3 major categories: cutaneous B-cell pseudolymphoma; cutaneous T-cell pseudolymphoma; and debatably lymphomatoid papulosis, a chronic, self-remitting, papulonecrotic condition that resembles lymphoma histologically but clinically appears benign. It is unknown if lymphomatoid papulosis represents a pseudolymphoma or a true lymphoma. Lymphomatoid papulosis may represent an early indolent form of CTCL.4

Pseudolymphomas can be triggered by a variety of causes. Most cases are idiopathic, and a causative stimulus is never identified. Drugs are known to cause many cases of pseudolymphoma, either by a causing a hypersensitivity reaction or by depressing immunosurveillance.5 Pseudolymphomas may result from exogenous stimuli such as jewelry, tattoo dyes, injectable fillers (eg, silicone), insect bites, vaccines, and trauma.6,7 Lastly, infections in the form of Borrelia, varicella, and molluscum contagiosum can potentially cause pseudolymphomas.4

Clinically, pseudolymphomas may demonstrate a B-cell or T-cell pattern. In cutaneous B-cell pseudolymphomas, asymptomatic solitary erythematous, violaceous, or flesh-colored nodules appear on the face, followed by the chest and arms. Cutaneous T-cell pseudolymphomas present with erythematous patches that are more likely to be symptomatic.4

Histologically, pseudolymphomas also are classified as demonstrating B-cell or T-cell patterns. The nodular inflammatory infiltrate of cutaneous B-cell pseudolymphoma corresponds with its clinically apparent nodules. It can be distinguished from lymphoma in that it is not solely a lymphocytic infiltrate but rather a mixed infiltrate including histiocytes, lymphocytes, eosinophils, and plasma cells. Additionally, cutaneous B-cell pseudolymphoma does not penetrate the dermis as deeply as CBCL.8 Cutaneous T-cell pseudolymphoma is more difficult to distinguish from CTCL because it also demonstrates a bandlike lymphocytic infiltrate in the papillary dermis with epidermotropism.9

Treatment must address the underlying cause of pseudolymphoma for resolution. Other treatment options include surgery, cryotherapy, local radiotherapy, topical steroids, and topical immunomodulators. Spontaneous resolution also can occur. The prognosis is better when a known trigger is eliminated, though idiopathic pseudolymphomas may be chronic in nature. It is important to rule out concurrent cutaneous lymphoma or rare transformation into cutaneous lymphoma.

Cutaneous T-Cell Lymphoma
Cutaneous T-cell lymphomas are a diverse group of neoplasms that account for most cutaneous lymphomas seen by dermatologists. In 1806, the first case of CTCL in the form of mycosis fungoides (MF) was described by Jean Louis Alibert. Mycosis fungoides represents the most common form of CTCL, accounting for approximately 50% of all primary cutaneous lymphomas.10 Mycosis fungoides was named after its morphological resemblance to mushrooms. Although not all cases exhibit a classic progression, MF is known for its stepwise progression from patch stage to tumor stage.

Clinically, lesions typically begin as patches that progress to plaques and finally tumors. This progression may not always occur and often can take years to decades to progress. Patches are characterized by erythematous, finely scaling lesions that may be easily confused with eczema or psoriasis. Lesions occur primarily in a swimming trunk distribution.

Mycosis fungoides histologically demonstrates a bandlike lymphocytic infiltrate with epidermotropism, which occurs when lymphocytes infiltrate the epidermis without spongiosis. These lymphocytes are larger, darker, and more angulated than normal lymphocytes. Intraepidermal nests of these atypical lymphocytes creating Pautrier microabscesses may be present. Tumor-stage lesions demonstrate diminished epidermotropism with dense sheets of lymphocytes in the dermis, and fat cells with cerebriform nuclei are present.

Therapies for MF may control the disease but may not prolong patients’ lives. Topical corticosteroids, phototherapy, and radiotherapy are options for skin-targeting therapies. Systemic chemotherapy and biological response modifiers also are viable treatment options. Prognosis for MF is poor.

There are a few notable variants of MF that are important to consider. Sézary syndrome is an erythrodermic variant of MF characterized by atypical Sézary cells. Clinically, it presents with generalized erythroderma with leonine facies, facial edema, and alopecia with associated symptoms of burning and pruritus. Histologically, Sézary syndrome is similar to MF with an increased CD4:CD8 ratio.10 Sézary syndrome may be treated with methotrexate or photopheresis, but the prognosis remains poor with an average survival of 5 years.

Cutaneous B-Cell Lymphoma
There are 5 types of primary CBCL: primary cutaneous follicle center lymphoma; primary cutaneous marginal zone B-cell lymphoma; primary cutaneous diffuse large B-cell lymphoma, other; precursor B-cell lymphoblastic lymphoma; and primary cutaneous DLBCLLT, which was seen in our patient.11

Primary cutaneous follicle center lymphoma is an indolent neoplastic proliferation in the skin. Clinically, it presents with solitary or grouped pinkish purple papules, plaques, or nodules on the trunk with surrounding patches of erythema.3 Lesions located on the back are referred to as Crosti lymphoma. Histopathology reveals a lymphocytic infiltrate with a diffuse follicular pattern and large round centroblasts, centrocytes, and immunoblasts with epidermal sparing. Tumor cells stain positively for κ or λ light chains, as well as CD20, CD79a, and B-cell lymphoma 6 (BCL-6); however, staining for the protein product of BCL-2 may be negative, which differentiates this form of CBCL from primary nodal B-cell lymphoma. Staining for MUM-1 may be negative, which contrasts with the strong expression seen in DLBCLLT. The follicular pattern of follicle center lymphoma stains positive for CD10, but the diffuse pattern may be CD10 negative. The prognosis for primary cutaneous follicle center lymphoma is favorable, but the recurrence rate is up to 50%.3 Treatment includes local radiotherapy or surgical excision.

Primary cutaneous marginal zone B-cell lymphoma is another indolent primary CBCL subtype that is closely related to mucosa-associated lymphoid tissue lymphomas and arises in areas of acrodermatitis chronica atrophicans and Borrelia infection. Clinically, it presents with recurrent, asymptomatic, red-brown papules, plaques, and nodules of the arms and legs. Histologically, there is a patchy infiltrate in the dermis and subcutis with sparing of the epidermis with pale-staining cells with indented nuclei, along with plasma cells and eosinophils. Primary cutaneous marginal zone B-cell lymphoma typically does not demonstrate epidermotropism. Centrocyte cells stain positively for CD20, CD79a, and BCL-2. The prognosis of primary cutaneous marginal zone B-cell lymphoma is favorable. Treatment is similar to primary cutaneous follicle center lymphoma with surgical excision, radiotherapy, and surveillance being the main modalities.

Primary cutaneous diffuse large B-cell lymphoma, other is an intermediately aggressive form of primary CBCL that is thought to be related to primary cutaneous DLBCLLT. Clinically, it presents with indurated erythematous to violaceous plaques on the trunk and thighs that may resemble a vascular tumor or panniculitis.2,12 Histopathologically, this form of lymphoma presents with a round cell morphology without BCL-2 expression, which distinguishes it from DLBCLLT. If limited to skin, the prognosis is better than the systemic form but is still less favorable than other forms of CBCL.

Precursor B-cell lymphoblastic lymphoma is an extremely rare type of CBCL that potentially can occur in the skin. It primarily affects children and young adults. Clinically, it presents as a solitary large erythematous tumor of the head. Histologically, monomorphic proliferation of medium-sized cells with round nuclei that demonstrate a starry sky pattern with multiple mitotic cells can be observed.13 Immunohistochemical markers such as CD43, CD10, CD20, and CD79a may be positive. The disease is very aggressive with poor prognosis if left untreated.

CONCLUSION

We present a rare case of primary cutaneous DLBCLLT. Our case demonstrates the classic presentation of primary cutaneous DLBCLLT in a 74-year-old woman with a tumor on the lower left leg. Histologically, a dense dermal and subcutis infiltrate of centroblasts and immunoblasts with a grenz zone was present. Immunostaining in our patient was consistent with characteristic findings in the literature, staining highly positive for BCL-2 and MUM-1. Primary cutaneous DLBCLLT is an extremely rare and unique form of cutaneous lymphoma that can have potentially fatal consequences if undiagnosed; therefore, clinicians must take great care to make the correct diagnosis based on a knowledge of the clinical and immunohistochemical findings of DLBCLLT.

References
  1. Sokol L, Naghashpour M, Glass LF. Primary cutaneous B-cell lymphomas: recent advances in diagnosis and management. Cancer Control. 2012;19:236-244.
  2. Grange F, Beylot-Barry M, Courville P, et al. Primary cutaneous diffuse large B-cell lymphoma, leg type: clinicopathologic features and prognostic analysis in 60 cases. Arch Dermatol. 2007;143:1144-1150.
  3. Willemze R, Jaffe ES, Burg G, et al. WHO-EORTC classification for cutaneous lymphomas. Blood. 2005;105:2768-3785.
  4. Brodell RT, Santa Cruz DJ. Cutaneous pseudolymphomas. Dermatol Clin. 1985;3:719-734.
  5. Albrecht J, Fine LA, Piette W. Drug-associated lymphoma and pseudolymphoma: recognition and management. Dermatol Clin. 2007;25:233-244; vii.
  6. Maubec E, Pinquier L, Viguier M, et al. Vaccination-induced cutaneous pseudolymphoma. J Am Acad Dermatol. 2005;52:623-629.
  7. Kluger N, Vermeulen C, Moguelet P, et al. Cutaneous lymphoid hyperplasia (pseudolymphoma) in tattoos: a case series of seven patients. J Eur Acad Dermatol Venereol. 2010;24:208-213.
  8. Burg G, Kerl H, Schmoeckel C. Differentiation between malignant B-cell lymphomas and pseudolymphomas of the skin. J Dermatol Surg Oncol. 1984;10:271-275.
  9. Ploysangam T, Breneman DL, Mutasim DF. Cutaneous pseudolymphomas. J Am Acad Dermatol. 1998;38(6, pt 1):877-895; quiz 896-897.
  10. Diamandidou E, Cohen PR, Kurzrock R. Mycosis fungoides and Sézary syndrome. Blood. 1996;88:2385-2409.
  11. Kempf W, Ralfkiaer E, Duncan LM, et al. Cutaneous marginal zone B-cell lymphoma. In: LeBoit P, Burg G, Weedon D, et al, eds. Pathology and Genetics of Skin Tumors. Lyon, France: IARC Press; 2006:194-195.
  12. Grange F, Bekkenk MW, Wechsler J, et al. Prognostic factors in cutaneous large B-cell lymphomas: a European multicentric study. J Clin Oncol. 2001;19:3602-3610.
  13. Chimenti S, Fink-Puches R, Peris K, et al. Cutaneous involvement in lymphoblastic lymphoma. J Cutan Pathol. 1999;26:379-385.
Article PDF
Author and Disclosure Information

All from Western University of Health Sciences, Pomona, California. Dr. Shitabata is from the Department of Dermatopathology. Drs. Athalye and Nami also are from the Pomona and College Medical Center, Long Beach, California.

The authors report no conflict of interest.

Correspondence: Leela Athalye, DO, 360 San Miguel Dr, Ste 501, Newport Beach, CA 92660 (calileela@gmail.com).

Issue
Cutis - 102(3)
Publications
Topics
Page Number
E31-E34
Sections
Author and Disclosure Information

All from Western University of Health Sciences, Pomona, California. Dr. Shitabata is from the Department of Dermatopathology. Drs. Athalye and Nami also are from the Pomona and College Medical Center, Long Beach, California.

The authors report no conflict of interest.

Correspondence: Leela Athalye, DO, 360 San Miguel Dr, Ste 501, Newport Beach, CA 92660 (calileela@gmail.com).

Author and Disclosure Information

All from Western University of Health Sciences, Pomona, California. Dr. Shitabata is from the Department of Dermatopathology. Drs. Athalye and Nami also are from the Pomona and College Medical Center, Long Beach, California.

The authors report no conflict of interest.

Correspondence: Leela Athalye, DO, 360 San Miguel Dr, Ste 501, Newport Beach, CA 92660 (calileela@gmail.com).

Article PDF
Article PDF

CASE REPORT

A 74-year-old woman presented with a painful lesion on the left lower leg that was getting larger and more edematous and erythematous over the last 5 months. She experienced numbness and burning of the left lower leg 1 year prior to the development of the lesion. A review of her medical history revealed an otherwise healthy woman with no constitutional symptoms of fever, chills, nausea, vomiting, diarrhea, or chest pain. The patient did not exhibit mucosal, genital, or nail involvement. Physical examination revealed a group of four 1-cm, ill-defined, irregularly bordered, violaceous plaques on the left anterior tibial leg with faint surrounding erythematous to violaceous patches (Figure 1). The plaques were tender to palpation with no bleeding or drainage.

Figure1
Figure 1. Primary cutaneous diffuse large B-cell lymphoma, leg type presenting as a violaceous, ill-defined, tender plaque with both surrounding and satellite faint erythematous to violaceous patches.

An 8.0-mm punch biopsy of the lesion was obtained. Hematoxylin and eosin staining on low-power magnification demonstrated a diffuse lymphocytic inflammatory infiltrate in the dermis and subcutis. Notable sparing of the subepidermal area (free grenz zone) was present (Figure 2A). On higher power, centroblasts and immunoblasts were visualized alongside extravasated red blood cells (Figure 2B). A diagnosis of primary cutaneous diffuse large B-cell lymphoma, leg type (DLBCLLT) was made. Various immunohistochemical stains confirmed the diagnosis, including B-cell lymphoma 2 (BCL-2)(Figure 3A) and multiple myeloma oncogene 1 (MUM-1)(Figure 3B), which were highly positive in our patient. The patient had a negative bone marrow biopsy and positron emission tomography scan. She was started on rituximab infusions and multiple radiation treatments. At 2-year follow-up the lymphoma continued to recur despite radiation therapy.

Figure2
Figure 2. Histopathology of primary cutaneous diffuse large B-cell lymphoma, leg type demonstrated a diffuse inflammatory infiltrate in the dermis and subcutis with a subepidermal free grenz zone on low power (A)(H&E). High-power view demonstrated centroblasts and immunoblasts alongside extravasated red blood cells (B)(H&E).

Figure 3. Positive B-cell lymphoma 2 (BCL-2)(A) and multiple myeloma oncogene 1 (MUM-1)(B) immunohistochemical staining confirmed the diagnosis.

COMMENT

Incidence and Clinical Characteristics

Primary cutaneous DLBCLLT is an intermediately aggressive form of primary cutaneous B-cell lymphoma (CBCL) that accounts for approximately 10% to 20% of all primary CBCLs and 1% to 3% of all cutaneous lymphomas.1 Diffuse large B-cell lymphoma, leg type primarily affects elderly patients (median age, 70 years). Women are more commonly affected. Clinically, primary cutaneous DLBCLLT presents as red-brown to bluish nodules or tumors on one or both distal legs. Although referred to as leg-type diffuse large B-cell lymphoma, 10% to 15% of patients have lesions in anatomic areas other than the legs, most commonly the trunk.

Histopathology

The diagnosis of DLBCLLT is best made histologically. There is a dense inflammatory infiltrate present in the dermis and subcutis that may extend upward into the dermoepidermal junction. Often a subepidermal free grenz zone may be seen, and adnexal structures may be destroyed. This infiltrate is composed of confluent sheets of large round cells including centroblasts and immunoblasts.2 Centroblasts are large cells that have nuclei with several small nucleoli adhering to the membrane, while immunoblasts are large round cells containing nuclei with large central nucleoli. Both centroblasts and immunoblasts stain positively for BCL-2. Centrocytes typically are absent. Staining for BCL-2 can be important in distinguishing DLBCLLT from other forms of CBCL. Diffuse large B-cell lymphoma, leg type also can demonstrate clusters of large atypical cells in the epidermis simulating epidermotropism and Pautrier microabscesses. Neoplastic cells in this condition may express monoclonal surface and cytoplasmic immunoglobulins. Primary cutaneous DLBCLLT typically is positive for B-cell markers CD20 and CD79a. Additionally, MUM-1/IRF4 (interferon regulatory factor 4) and forkhead box protein 1 (FOXP1) are strongly expressed by most patients, which helps distinguish it from other forms of CBCL.

Treatment

Diffuse large B-cell lymphoma, leg type is a relatively aggressive form of CBCL that requires more aggressive treatment than the conservative watchful waiting of some of the more indolent forms of primary CBCL. One regimen involves using cyclophosphamide, doxorubicin, vincristine, and prednisone plus rituximab. Local chemotherapy or radiation with rituximab is another treatment option.1,2 In patients with severe comorbidities, rituximab alone may be administered. The prognosis for DLBCLLT is not as favorable as other types of primary CBCL, with an estimated 5-year survival rate of approximately 50%.2

 

 

Differential Diagnosis

Lymphomas are malignancies of the lymphocytes that may be subdivided depending on the organ of origin. Both primary nodal lymphomas and primary cutaneous lymphomas exist. Primary nodal lymphomas arise from the lymph nodes and are divided into Hodgkin and non-Hodgkin lymphomas. There are 2 major types of primary cutaneous lymphomas: cutaneous T-cell lymphoma (CTCL) and CBCL. Most primary cutaneous lymphomas are CTCLs, accounting for 75% to 80%.3

Pseudolymphoma
Pseudolymphoma is an inflammatory condition that may histologically mimic cutaneous lymphoma but has a benign clinical course. Pseudolymphoma is not a specific disease but rather is a reactive lymphoproliferative response to a known or unknown stimulus.4 Pseudolymphoma can be broken down into 2 or 3 major categories: cutaneous B-cell pseudolymphoma; cutaneous T-cell pseudolymphoma; and debatably lymphomatoid papulosis, a chronic, self-remitting, papulonecrotic condition that resembles lymphoma histologically but clinically appears benign. It is unknown if lymphomatoid papulosis represents a pseudolymphoma or a true lymphoma. Lymphomatoid papulosis may represent an early indolent form of CTCL.4

Pseudolymphomas can be triggered by a variety of causes. Most cases are idiopathic, and a causative stimulus is never identified. Drugs are known to cause many cases of pseudolymphoma, either by a causing a hypersensitivity reaction or by depressing immunosurveillance.5 Pseudolymphomas may result from exogenous stimuli such as jewelry, tattoo dyes, injectable fillers (eg, silicone), insect bites, vaccines, and trauma.6,7 Lastly, infections in the form of Borrelia, varicella, and molluscum contagiosum can potentially cause pseudolymphomas.4

Clinically, pseudolymphomas may demonstrate a B-cell or T-cell pattern. In cutaneous B-cell pseudolymphomas, asymptomatic solitary erythematous, violaceous, or flesh-colored nodules appear on the face, followed by the chest and arms. Cutaneous T-cell pseudolymphomas present with erythematous patches that are more likely to be symptomatic.4

Histologically, pseudolymphomas also are classified as demonstrating B-cell or T-cell patterns. The nodular inflammatory infiltrate of cutaneous B-cell pseudolymphoma corresponds with its clinically apparent nodules. It can be distinguished from lymphoma in that it is not solely a lymphocytic infiltrate but rather a mixed infiltrate including histiocytes, lymphocytes, eosinophils, and plasma cells. Additionally, cutaneous B-cell pseudolymphoma does not penetrate the dermis as deeply as CBCL.8 Cutaneous T-cell pseudolymphoma is more difficult to distinguish from CTCL because it also demonstrates a bandlike lymphocytic infiltrate in the papillary dermis with epidermotropism.9

Treatment must address the underlying cause of pseudolymphoma for resolution. Other treatment options include surgery, cryotherapy, local radiotherapy, topical steroids, and topical immunomodulators. Spontaneous resolution also can occur. The prognosis is better when a known trigger is eliminated, though idiopathic pseudolymphomas may be chronic in nature. It is important to rule out concurrent cutaneous lymphoma or rare transformation into cutaneous lymphoma.

Cutaneous T-Cell Lymphoma
Cutaneous T-cell lymphomas are a diverse group of neoplasms that account for most cutaneous lymphomas seen by dermatologists. In 1806, the first case of CTCL in the form of mycosis fungoides (MF) was described by Jean Louis Alibert. Mycosis fungoides represents the most common form of CTCL, accounting for approximately 50% of all primary cutaneous lymphomas.10 Mycosis fungoides was named after its morphological resemblance to mushrooms. Although not all cases exhibit a classic progression, MF is known for its stepwise progression from patch stage to tumor stage.

Clinically, lesions typically begin as patches that progress to plaques and finally tumors. This progression may not always occur and often can take years to decades to progress. Patches are characterized by erythematous, finely scaling lesions that may be easily confused with eczema or psoriasis. Lesions occur primarily in a swimming trunk distribution.

Mycosis fungoides histologically demonstrates a bandlike lymphocytic infiltrate with epidermotropism, which occurs when lymphocytes infiltrate the epidermis without spongiosis. These lymphocytes are larger, darker, and more angulated than normal lymphocytes. Intraepidermal nests of these atypical lymphocytes creating Pautrier microabscesses may be present. Tumor-stage lesions demonstrate diminished epidermotropism with dense sheets of lymphocytes in the dermis, and fat cells with cerebriform nuclei are present.

Therapies for MF may control the disease but may not prolong patients’ lives. Topical corticosteroids, phototherapy, and radiotherapy are options for skin-targeting therapies. Systemic chemotherapy and biological response modifiers also are viable treatment options. Prognosis for MF is poor.

There are a few notable variants of MF that are important to consider. Sézary syndrome is an erythrodermic variant of MF characterized by atypical Sézary cells. Clinically, it presents with generalized erythroderma with leonine facies, facial edema, and alopecia with associated symptoms of burning and pruritus. Histologically, Sézary syndrome is similar to MF with an increased CD4:CD8 ratio.10 Sézary syndrome may be treated with methotrexate or photopheresis, but the prognosis remains poor with an average survival of 5 years.

Cutaneous B-Cell Lymphoma
There are 5 types of primary CBCL: primary cutaneous follicle center lymphoma; primary cutaneous marginal zone B-cell lymphoma; primary cutaneous diffuse large B-cell lymphoma, other; precursor B-cell lymphoblastic lymphoma; and primary cutaneous DLBCLLT, which was seen in our patient.11

Primary cutaneous follicle center lymphoma is an indolent neoplastic proliferation in the skin. Clinically, it presents with solitary or grouped pinkish purple papules, plaques, or nodules on the trunk with surrounding patches of erythema.3 Lesions located on the back are referred to as Crosti lymphoma. Histopathology reveals a lymphocytic infiltrate with a diffuse follicular pattern and large round centroblasts, centrocytes, and immunoblasts with epidermal sparing. Tumor cells stain positively for κ or λ light chains, as well as CD20, CD79a, and B-cell lymphoma 6 (BCL-6); however, staining for the protein product of BCL-2 may be negative, which differentiates this form of CBCL from primary nodal B-cell lymphoma. Staining for MUM-1 may be negative, which contrasts with the strong expression seen in DLBCLLT. The follicular pattern of follicle center lymphoma stains positive for CD10, but the diffuse pattern may be CD10 negative. The prognosis for primary cutaneous follicle center lymphoma is favorable, but the recurrence rate is up to 50%.3 Treatment includes local radiotherapy or surgical excision.

Primary cutaneous marginal zone B-cell lymphoma is another indolent primary CBCL subtype that is closely related to mucosa-associated lymphoid tissue lymphomas and arises in areas of acrodermatitis chronica atrophicans and Borrelia infection. Clinically, it presents with recurrent, asymptomatic, red-brown papules, plaques, and nodules of the arms and legs. Histologically, there is a patchy infiltrate in the dermis and subcutis with sparing of the epidermis with pale-staining cells with indented nuclei, along with plasma cells and eosinophils. Primary cutaneous marginal zone B-cell lymphoma typically does not demonstrate epidermotropism. Centrocyte cells stain positively for CD20, CD79a, and BCL-2. The prognosis of primary cutaneous marginal zone B-cell lymphoma is favorable. Treatment is similar to primary cutaneous follicle center lymphoma with surgical excision, radiotherapy, and surveillance being the main modalities.

Primary cutaneous diffuse large B-cell lymphoma, other is an intermediately aggressive form of primary CBCL that is thought to be related to primary cutaneous DLBCLLT. Clinically, it presents with indurated erythematous to violaceous plaques on the trunk and thighs that may resemble a vascular tumor or panniculitis.2,12 Histopathologically, this form of lymphoma presents with a round cell morphology without BCL-2 expression, which distinguishes it from DLBCLLT. If limited to skin, the prognosis is better than the systemic form but is still less favorable than other forms of CBCL.

Precursor B-cell lymphoblastic lymphoma is an extremely rare type of CBCL that potentially can occur in the skin. It primarily affects children and young adults. Clinically, it presents as a solitary large erythematous tumor of the head. Histologically, monomorphic proliferation of medium-sized cells with round nuclei that demonstrate a starry sky pattern with multiple mitotic cells can be observed.13 Immunohistochemical markers such as CD43, CD10, CD20, and CD79a may be positive. The disease is very aggressive with poor prognosis if left untreated.

CONCLUSION

We present a rare case of primary cutaneous DLBCLLT. Our case demonstrates the classic presentation of primary cutaneous DLBCLLT in a 74-year-old woman with a tumor on the lower left leg. Histologically, a dense dermal and subcutis infiltrate of centroblasts and immunoblasts with a grenz zone was present. Immunostaining in our patient was consistent with characteristic findings in the literature, staining highly positive for BCL-2 and MUM-1. Primary cutaneous DLBCLLT is an extremely rare and unique form of cutaneous lymphoma that can have potentially fatal consequences if undiagnosed; therefore, clinicians must take great care to make the correct diagnosis based on a knowledge of the clinical and immunohistochemical findings of DLBCLLT.

CASE REPORT

A 74-year-old woman presented with a painful lesion on the left lower leg that was getting larger and more edematous and erythematous over the last 5 months. She experienced numbness and burning of the left lower leg 1 year prior to the development of the lesion. A review of her medical history revealed an otherwise healthy woman with no constitutional symptoms of fever, chills, nausea, vomiting, diarrhea, or chest pain. The patient did not exhibit mucosal, genital, or nail involvement. Physical examination revealed a group of four 1-cm, ill-defined, irregularly bordered, violaceous plaques on the left anterior tibial leg with faint surrounding erythematous to violaceous patches (Figure 1). The plaques were tender to palpation with no bleeding or drainage.

Figure1
Figure 1. Primary cutaneous diffuse large B-cell lymphoma, leg type presenting as a violaceous, ill-defined, tender plaque with both surrounding and satellite faint erythematous to violaceous patches.

An 8.0-mm punch biopsy of the lesion was obtained. Hematoxylin and eosin staining on low-power magnification demonstrated a diffuse lymphocytic inflammatory infiltrate in the dermis and subcutis. Notable sparing of the subepidermal area (free grenz zone) was present (Figure 2A). On higher power, centroblasts and immunoblasts were visualized alongside extravasated red blood cells (Figure 2B). A diagnosis of primary cutaneous diffuse large B-cell lymphoma, leg type (DLBCLLT) was made. Various immunohistochemical stains confirmed the diagnosis, including B-cell lymphoma 2 (BCL-2)(Figure 3A) and multiple myeloma oncogene 1 (MUM-1)(Figure 3B), which were highly positive in our patient. The patient had a negative bone marrow biopsy and positron emission tomography scan. She was started on rituximab infusions and multiple radiation treatments. At 2-year follow-up the lymphoma continued to recur despite radiation therapy.

Figure2
Figure 2. Histopathology of primary cutaneous diffuse large B-cell lymphoma, leg type demonstrated a diffuse inflammatory infiltrate in the dermis and subcutis with a subepidermal free grenz zone on low power (A)(H&E). High-power view demonstrated centroblasts and immunoblasts alongside extravasated red blood cells (B)(H&E).

Figure 3. Positive B-cell lymphoma 2 (BCL-2)(A) and multiple myeloma oncogene 1 (MUM-1)(B) immunohistochemical staining confirmed the diagnosis.

COMMENT

Incidence and Clinical Characteristics

Primary cutaneous DLBCLLT is an intermediately aggressive form of primary cutaneous B-cell lymphoma (CBCL) that accounts for approximately 10% to 20% of all primary CBCLs and 1% to 3% of all cutaneous lymphomas.1 Diffuse large B-cell lymphoma, leg type primarily affects elderly patients (median age, 70 years). Women are more commonly affected. Clinically, primary cutaneous DLBCLLT presents as red-brown to bluish nodules or tumors on one or both distal legs. Although referred to as leg-type diffuse large B-cell lymphoma, 10% to 15% of patients have lesions in anatomic areas other than the legs, most commonly the trunk.

Histopathology

The diagnosis of DLBCLLT is best made histologically. There is a dense inflammatory infiltrate present in the dermis and subcutis that may extend upward into the dermoepidermal junction. Often a subepidermal free grenz zone may be seen, and adnexal structures may be destroyed. This infiltrate is composed of confluent sheets of large round cells including centroblasts and immunoblasts.2 Centroblasts are large cells that have nuclei with several small nucleoli adhering to the membrane, while immunoblasts are large round cells containing nuclei with large central nucleoli. Both centroblasts and immunoblasts stain positively for BCL-2. Centrocytes typically are absent. Staining for BCL-2 can be important in distinguishing DLBCLLT from other forms of CBCL. Diffuse large B-cell lymphoma, leg type also can demonstrate clusters of large atypical cells in the epidermis simulating epidermotropism and Pautrier microabscesses. Neoplastic cells in this condition may express monoclonal surface and cytoplasmic immunoglobulins. Primary cutaneous DLBCLLT typically is positive for B-cell markers CD20 and CD79a. Additionally, MUM-1/IRF4 (interferon regulatory factor 4) and forkhead box protein 1 (FOXP1) are strongly expressed by most patients, which helps distinguish it from other forms of CBCL.

Treatment

Diffuse large B-cell lymphoma, leg type is a relatively aggressive form of CBCL that requires more aggressive treatment than the conservative watchful waiting of some of the more indolent forms of primary CBCL. One regimen involves using cyclophosphamide, doxorubicin, vincristine, and prednisone plus rituximab. Local chemotherapy or radiation with rituximab is another treatment option.1,2 In patients with severe comorbidities, rituximab alone may be administered. The prognosis for DLBCLLT is not as favorable as other types of primary CBCL, with an estimated 5-year survival rate of approximately 50%.2

 

 

Differential Diagnosis

Lymphomas are malignancies of the lymphocytes that may be subdivided depending on the organ of origin. Both primary nodal lymphomas and primary cutaneous lymphomas exist. Primary nodal lymphomas arise from the lymph nodes and are divided into Hodgkin and non-Hodgkin lymphomas. There are 2 major types of primary cutaneous lymphomas: cutaneous T-cell lymphoma (CTCL) and CBCL. Most primary cutaneous lymphomas are CTCLs, accounting for 75% to 80%.3

Pseudolymphoma
Pseudolymphoma is an inflammatory condition that may histologically mimic cutaneous lymphoma but has a benign clinical course. Pseudolymphoma is not a specific disease but rather is a reactive lymphoproliferative response to a known or unknown stimulus.4 Pseudolymphoma can be broken down into 2 or 3 major categories: cutaneous B-cell pseudolymphoma; cutaneous T-cell pseudolymphoma; and debatably lymphomatoid papulosis, a chronic, self-remitting, papulonecrotic condition that resembles lymphoma histologically but clinically appears benign. It is unknown if lymphomatoid papulosis represents a pseudolymphoma or a true lymphoma. Lymphomatoid papulosis may represent an early indolent form of CTCL.4

Pseudolymphomas can be triggered by a variety of causes. Most cases are idiopathic, and a causative stimulus is never identified. Drugs are known to cause many cases of pseudolymphoma, either by a causing a hypersensitivity reaction or by depressing immunosurveillance.5 Pseudolymphomas may result from exogenous stimuli such as jewelry, tattoo dyes, injectable fillers (eg, silicone), insect bites, vaccines, and trauma.6,7 Lastly, infections in the form of Borrelia, varicella, and molluscum contagiosum can potentially cause pseudolymphomas.4

Clinically, pseudolymphomas may demonstrate a B-cell or T-cell pattern. In cutaneous B-cell pseudolymphomas, asymptomatic solitary erythematous, violaceous, or flesh-colored nodules appear on the face, followed by the chest and arms. Cutaneous T-cell pseudolymphomas present with erythematous patches that are more likely to be symptomatic.4

Histologically, pseudolymphomas also are classified as demonstrating B-cell or T-cell patterns. The nodular inflammatory infiltrate of cutaneous B-cell pseudolymphoma corresponds with its clinically apparent nodules. It can be distinguished from lymphoma in that it is not solely a lymphocytic infiltrate but rather a mixed infiltrate including histiocytes, lymphocytes, eosinophils, and plasma cells. Additionally, cutaneous B-cell pseudolymphoma does not penetrate the dermis as deeply as CBCL.8 Cutaneous T-cell pseudolymphoma is more difficult to distinguish from CTCL because it also demonstrates a bandlike lymphocytic infiltrate in the papillary dermis with epidermotropism.9

Treatment must address the underlying cause of pseudolymphoma for resolution. Other treatment options include surgery, cryotherapy, local radiotherapy, topical steroids, and topical immunomodulators. Spontaneous resolution also can occur. The prognosis is better when a known trigger is eliminated, though idiopathic pseudolymphomas may be chronic in nature. It is important to rule out concurrent cutaneous lymphoma or rare transformation into cutaneous lymphoma.

Cutaneous T-Cell Lymphoma
Cutaneous T-cell lymphomas are a diverse group of neoplasms that account for most cutaneous lymphomas seen by dermatologists. In 1806, the first case of CTCL in the form of mycosis fungoides (MF) was described by Jean Louis Alibert. Mycosis fungoides represents the most common form of CTCL, accounting for approximately 50% of all primary cutaneous lymphomas.10 Mycosis fungoides was named after its morphological resemblance to mushrooms. Although not all cases exhibit a classic progression, MF is known for its stepwise progression from patch stage to tumor stage.

Clinically, lesions typically begin as patches that progress to plaques and finally tumors. This progression may not always occur and often can take years to decades to progress. Patches are characterized by erythematous, finely scaling lesions that may be easily confused with eczema or psoriasis. Lesions occur primarily in a swimming trunk distribution.

Mycosis fungoides histologically demonstrates a bandlike lymphocytic infiltrate with epidermotropism, which occurs when lymphocytes infiltrate the epidermis without spongiosis. These lymphocytes are larger, darker, and more angulated than normal lymphocytes. Intraepidermal nests of these atypical lymphocytes creating Pautrier microabscesses may be present. Tumor-stage lesions demonstrate diminished epidermotropism with dense sheets of lymphocytes in the dermis, and fat cells with cerebriform nuclei are present.

Therapies for MF may control the disease but may not prolong patients’ lives. Topical corticosteroids, phototherapy, and radiotherapy are options for skin-targeting therapies. Systemic chemotherapy and biological response modifiers also are viable treatment options. Prognosis for MF is poor.

There are a few notable variants of MF that are important to consider. Sézary syndrome is an erythrodermic variant of MF characterized by atypical Sézary cells. Clinically, it presents with generalized erythroderma with leonine facies, facial edema, and alopecia with associated symptoms of burning and pruritus. Histologically, Sézary syndrome is similar to MF with an increased CD4:CD8 ratio.10 Sézary syndrome may be treated with methotrexate or photopheresis, but the prognosis remains poor with an average survival of 5 years.

Cutaneous B-Cell Lymphoma
There are 5 types of primary CBCL: primary cutaneous follicle center lymphoma; primary cutaneous marginal zone B-cell lymphoma; primary cutaneous diffuse large B-cell lymphoma, other; precursor B-cell lymphoblastic lymphoma; and primary cutaneous DLBCLLT, which was seen in our patient.11

Primary cutaneous follicle center lymphoma is an indolent neoplastic proliferation in the skin. Clinically, it presents with solitary or grouped pinkish purple papules, plaques, or nodules on the trunk with surrounding patches of erythema.3 Lesions located on the back are referred to as Crosti lymphoma. Histopathology reveals a lymphocytic infiltrate with a diffuse follicular pattern and large round centroblasts, centrocytes, and immunoblasts with epidermal sparing. Tumor cells stain positively for κ or λ light chains, as well as CD20, CD79a, and B-cell lymphoma 6 (BCL-6); however, staining for the protein product of BCL-2 may be negative, which differentiates this form of CBCL from primary nodal B-cell lymphoma. Staining for MUM-1 may be negative, which contrasts with the strong expression seen in DLBCLLT. The follicular pattern of follicle center lymphoma stains positive for CD10, but the diffuse pattern may be CD10 negative. The prognosis for primary cutaneous follicle center lymphoma is favorable, but the recurrence rate is up to 50%.3 Treatment includes local radiotherapy or surgical excision.

Primary cutaneous marginal zone B-cell lymphoma is another indolent primary CBCL subtype that is closely related to mucosa-associated lymphoid tissue lymphomas and arises in areas of acrodermatitis chronica atrophicans and Borrelia infection. Clinically, it presents with recurrent, asymptomatic, red-brown papules, plaques, and nodules of the arms and legs. Histologically, there is a patchy infiltrate in the dermis and subcutis with sparing of the epidermis with pale-staining cells with indented nuclei, along with plasma cells and eosinophils. Primary cutaneous marginal zone B-cell lymphoma typically does not demonstrate epidermotropism. Centrocyte cells stain positively for CD20, CD79a, and BCL-2. The prognosis of primary cutaneous marginal zone B-cell lymphoma is favorable. Treatment is similar to primary cutaneous follicle center lymphoma with surgical excision, radiotherapy, and surveillance being the main modalities.

Primary cutaneous diffuse large B-cell lymphoma, other is an intermediately aggressive form of primary CBCL that is thought to be related to primary cutaneous DLBCLLT. Clinically, it presents with indurated erythematous to violaceous plaques on the trunk and thighs that may resemble a vascular tumor or panniculitis.2,12 Histopathologically, this form of lymphoma presents with a round cell morphology without BCL-2 expression, which distinguishes it from DLBCLLT. If limited to skin, the prognosis is better than the systemic form but is still less favorable than other forms of CBCL.

Precursor B-cell lymphoblastic lymphoma is an extremely rare type of CBCL that potentially can occur in the skin. It primarily affects children and young adults. Clinically, it presents as a solitary large erythematous tumor of the head. Histologically, monomorphic proliferation of medium-sized cells with round nuclei that demonstrate a starry sky pattern with multiple mitotic cells can be observed.13 Immunohistochemical markers such as CD43, CD10, CD20, and CD79a may be positive. The disease is very aggressive with poor prognosis if left untreated.

CONCLUSION

We present a rare case of primary cutaneous DLBCLLT. Our case demonstrates the classic presentation of primary cutaneous DLBCLLT in a 74-year-old woman with a tumor on the lower left leg. Histologically, a dense dermal and subcutis infiltrate of centroblasts and immunoblasts with a grenz zone was present. Immunostaining in our patient was consistent with characteristic findings in the literature, staining highly positive for BCL-2 and MUM-1. Primary cutaneous DLBCLLT is an extremely rare and unique form of cutaneous lymphoma that can have potentially fatal consequences if undiagnosed; therefore, clinicians must take great care to make the correct diagnosis based on a knowledge of the clinical and immunohistochemical findings of DLBCLLT.

References
  1. Sokol L, Naghashpour M, Glass LF. Primary cutaneous B-cell lymphomas: recent advances in diagnosis and management. Cancer Control. 2012;19:236-244.
  2. Grange F, Beylot-Barry M, Courville P, et al. Primary cutaneous diffuse large B-cell lymphoma, leg type: clinicopathologic features and prognostic analysis in 60 cases. Arch Dermatol. 2007;143:1144-1150.
  3. Willemze R, Jaffe ES, Burg G, et al. WHO-EORTC classification for cutaneous lymphomas. Blood. 2005;105:2768-3785.
  4. Brodell RT, Santa Cruz DJ. Cutaneous pseudolymphomas. Dermatol Clin. 1985;3:719-734.
  5. Albrecht J, Fine LA, Piette W. Drug-associated lymphoma and pseudolymphoma: recognition and management. Dermatol Clin. 2007;25:233-244; vii.
  6. Maubec E, Pinquier L, Viguier M, et al. Vaccination-induced cutaneous pseudolymphoma. J Am Acad Dermatol. 2005;52:623-629.
  7. Kluger N, Vermeulen C, Moguelet P, et al. Cutaneous lymphoid hyperplasia (pseudolymphoma) in tattoos: a case series of seven patients. J Eur Acad Dermatol Venereol. 2010;24:208-213.
  8. Burg G, Kerl H, Schmoeckel C. Differentiation between malignant B-cell lymphomas and pseudolymphomas of the skin. J Dermatol Surg Oncol. 1984;10:271-275.
  9. Ploysangam T, Breneman DL, Mutasim DF. Cutaneous pseudolymphomas. J Am Acad Dermatol. 1998;38(6, pt 1):877-895; quiz 896-897.
  10. Diamandidou E, Cohen PR, Kurzrock R. Mycosis fungoides and Sézary syndrome. Blood. 1996;88:2385-2409.
  11. Kempf W, Ralfkiaer E, Duncan LM, et al. Cutaneous marginal zone B-cell lymphoma. In: LeBoit P, Burg G, Weedon D, et al, eds. Pathology and Genetics of Skin Tumors. Lyon, France: IARC Press; 2006:194-195.
  12. Grange F, Bekkenk MW, Wechsler J, et al. Prognostic factors in cutaneous large B-cell lymphomas: a European multicentric study. J Clin Oncol. 2001;19:3602-3610.
  13. Chimenti S, Fink-Puches R, Peris K, et al. Cutaneous involvement in lymphoblastic lymphoma. J Cutan Pathol. 1999;26:379-385.
References
  1. Sokol L, Naghashpour M, Glass LF. Primary cutaneous B-cell lymphomas: recent advances in diagnosis and management. Cancer Control. 2012;19:236-244.
  2. Grange F, Beylot-Barry M, Courville P, et al. Primary cutaneous diffuse large B-cell lymphoma, leg type: clinicopathologic features and prognostic analysis in 60 cases. Arch Dermatol. 2007;143:1144-1150.
  3. Willemze R, Jaffe ES, Burg G, et al. WHO-EORTC classification for cutaneous lymphomas. Blood. 2005;105:2768-3785.
  4. Brodell RT, Santa Cruz DJ. Cutaneous pseudolymphomas. Dermatol Clin. 1985;3:719-734.
  5. Albrecht J, Fine LA, Piette W. Drug-associated lymphoma and pseudolymphoma: recognition and management. Dermatol Clin. 2007;25:233-244; vii.
  6. Maubec E, Pinquier L, Viguier M, et al. Vaccination-induced cutaneous pseudolymphoma. J Am Acad Dermatol. 2005;52:623-629.
  7. Kluger N, Vermeulen C, Moguelet P, et al. Cutaneous lymphoid hyperplasia (pseudolymphoma) in tattoos: a case series of seven patients. J Eur Acad Dermatol Venereol. 2010;24:208-213.
  8. Burg G, Kerl H, Schmoeckel C. Differentiation between malignant B-cell lymphomas and pseudolymphomas of the skin. J Dermatol Surg Oncol. 1984;10:271-275.
  9. Ploysangam T, Breneman DL, Mutasim DF. Cutaneous pseudolymphomas. J Am Acad Dermatol. 1998;38(6, pt 1):877-895; quiz 896-897.
  10. Diamandidou E, Cohen PR, Kurzrock R. Mycosis fungoides and Sézary syndrome. Blood. 1996;88:2385-2409.
  11. Kempf W, Ralfkiaer E, Duncan LM, et al. Cutaneous marginal zone B-cell lymphoma. In: LeBoit P, Burg G, Weedon D, et al, eds. Pathology and Genetics of Skin Tumors. Lyon, France: IARC Press; 2006:194-195.
  12. Grange F, Bekkenk MW, Wechsler J, et al. Prognostic factors in cutaneous large B-cell lymphomas: a European multicentric study. J Clin Oncol. 2001;19:3602-3610.
  13. Chimenti S, Fink-Puches R, Peris K, et al. Cutaneous involvement in lymphoblastic lymphoma. J Cutan Pathol. 1999;26:379-385.
Issue
Cutis - 102(3)
Issue
Cutis - 102(3)
Page Number
E31-E34
Page Number
E31-E34
Publications
Publications
Topics
Article Type
Sections
Inside the Article

Practice Points

  • Primary cutaneous diffuse large B-cell lymphoma, leg type (DLBCLLT) is characterized by the presence of large round cells on histopathology.
  • There are potentially fatal consequences if undiagnosed; therefore, clinicians must take great care to make the correct diagnosis based on a knowledge of the clinical and immunohistochemical findings of DLBCLLT.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Article PDF Media

Itraconazole for BCC: Does it work?

Article Type
Changed
Fri, 01/18/2019 - 17:55

 

Oral itraconazole shows potential for treatment of locally advanced or metastatic basal cell carcinoma (BCC) as well as Gorlin syndrome, Justin J. Leitenberger, MD, declared at the annual meeting of the American College of Mohs Surgery.

Bruce Jancin/MDedge News
Dr. Justin J. Leitenberger

The oral antifungal inhibits the hedgehog signaling pathway, a key driver of BCC. And while itraconazole is not as potent an inhibitor of hedgehog pathway expression as, say vismodegib (Erivedge), the antifungal acts at a separate site on the pathway, making it an attractive candidate for combination therapy, explained Dr. Leitenberger of Oregon Health & Science University, Portland.

Dermatologists at Stanford University have led the way in exploring oral itraconazole as a treatment for BCC. Among a series of 29 patients with one or more large but nonadvanced BCCs, 19 were treated using oral itraconazole at 200 mg b.i.d. for 1 month or 100 mg b.i.d. for an average of 2.3 months. Hedgehog pathway expression was reduced by 65% in the itraconazole-treated patients, as compared with the 90% reduction which is achieved with vismodegib.

Of more direct clinical relevance, however, itraconazole also reduced tumor area by 24%. Four of eight patients with 57 tumors achieved a partial response, and the other four had stable disease (J Clin Oncol. 2014 Mar 10;32[8]:745-51).

The Stanford group has also shown that combination therapy with oral itraconazole and intravenous arsenic trioxide reduces hedgehog pathway expression by 75%, up by an absolute 10% from itraconazole alone. The two agents inhibit the pathway at different sites.


Five patients with metastatic BCC who relapsed after treatment with vismodegib received intravenous arsenic trioxide for the first 5 days of every month, followed by oral itraconazole at 200 mg b.i.d. on days 6-28. Three patients completed three such cycles, while two discontinued early because of progressive disease or adverse events including a grade 3 infection and grade 2 transaminitis. All three patients who completed three treatment cycles achieved stable disease. The Stanford investigators speculated that concurrent continuous dosing might be required to obtain tumor shrinkage (JAMA Dermatol. 2016 Apr;152[4]:452-6).

Lots more work remains to be done in order to optimize combination therapy utilizing oral itraconazole for advanced BCC. Different dosing regimens and combinations of hedgehog pathway inhibitors need to be studied. Another important question is how effective itraconazole-based combinations will be in vismodegib-naive as compared with vismodegib-resistant patients, Dr. Leitenberger observed.

He reported having no financial conflicts regarding his presentation.

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

 

Oral itraconazole shows potential for treatment of locally advanced or metastatic basal cell carcinoma (BCC) as well as Gorlin syndrome, Justin J. Leitenberger, MD, declared at the annual meeting of the American College of Mohs Surgery.

Bruce Jancin/MDedge News
Dr. Justin J. Leitenberger

The oral antifungal inhibits the hedgehog signaling pathway, a key driver of BCC. And while itraconazole is not as potent an inhibitor of hedgehog pathway expression as, say vismodegib (Erivedge), the antifungal acts at a separate site on the pathway, making it an attractive candidate for combination therapy, explained Dr. Leitenberger of Oregon Health & Science University, Portland.

Dermatologists at Stanford University have led the way in exploring oral itraconazole as a treatment for BCC. Among a series of 29 patients with one or more large but nonadvanced BCCs, 19 were treated using oral itraconazole at 200 mg b.i.d. for 1 month or 100 mg b.i.d. for an average of 2.3 months. Hedgehog pathway expression was reduced by 65% in the itraconazole-treated patients, as compared with the 90% reduction which is achieved with vismodegib.

Of more direct clinical relevance, however, itraconazole also reduced tumor area by 24%. Four of eight patients with 57 tumors achieved a partial response, and the other four had stable disease (J Clin Oncol. 2014 Mar 10;32[8]:745-51).

The Stanford group has also shown that combination therapy with oral itraconazole and intravenous arsenic trioxide reduces hedgehog pathway expression by 75%, up by an absolute 10% from itraconazole alone. The two agents inhibit the pathway at different sites.


Five patients with metastatic BCC who relapsed after treatment with vismodegib received intravenous arsenic trioxide for the first 5 days of every month, followed by oral itraconazole at 200 mg b.i.d. on days 6-28. Three patients completed three such cycles, while two discontinued early because of progressive disease or adverse events including a grade 3 infection and grade 2 transaminitis. All three patients who completed three treatment cycles achieved stable disease. The Stanford investigators speculated that concurrent continuous dosing might be required to obtain tumor shrinkage (JAMA Dermatol. 2016 Apr;152[4]:452-6).

Lots more work remains to be done in order to optimize combination therapy utilizing oral itraconazole for advanced BCC. Different dosing regimens and combinations of hedgehog pathway inhibitors need to be studied. Another important question is how effective itraconazole-based combinations will be in vismodegib-naive as compared with vismodegib-resistant patients, Dr. Leitenberger observed.

He reported having no financial conflicts regarding his presentation.

 

Oral itraconazole shows potential for treatment of locally advanced or metastatic basal cell carcinoma (BCC) as well as Gorlin syndrome, Justin J. Leitenberger, MD, declared at the annual meeting of the American College of Mohs Surgery.

Bruce Jancin/MDedge News
Dr. Justin J. Leitenberger

The oral antifungal inhibits the hedgehog signaling pathway, a key driver of BCC. And while itraconazole is not as potent an inhibitor of hedgehog pathway expression as, say vismodegib (Erivedge), the antifungal acts at a separate site on the pathway, making it an attractive candidate for combination therapy, explained Dr. Leitenberger of Oregon Health & Science University, Portland.

Dermatologists at Stanford University have led the way in exploring oral itraconazole as a treatment for BCC. Among a series of 29 patients with one or more large but nonadvanced BCCs, 19 were treated using oral itraconazole at 200 mg b.i.d. for 1 month or 100 mg b.i.d. for an average of 2.3 months. Hedgehog pathway expression was reduced by 65% in the itraconazole-treated patients, as compared with the 90% reduction which is achieved with vismodegib.

Of more direct clinical relevance, however, itraconazole also reduced tumor area by 24%. Four of eight patients with 57 tumors achieved a partial response, and the other four had stable disease (J Clin Oncol. 2014 Mar 10;32[8]:745-51).

The Stanford group has also shown that combination therapy with oral itraconazole and intravenous arsenic trioxide reduces hedgehog pathway expression by 75%, up by an absolute 10% from itraconazole alone. The two agents inhibit the pathway at different sites.


Five patients with metastatic BCC who relapsed after treatment with vismodegib received intravenous arsenic trioxide for the first 5 days of every month, followed by oral itraconazole at 200 mg b.i.d. on days 6-28. Three patients completed three such cycles, while two discontinued early because of progressive disease or adverse events including a grade 3 infection and grade 2 transaminitis. All three patients who completed three treatment cycles achieved stable disease. The Stanford investigators speculated that concurrent continuous dosing might be required to obtain tumor shrinkage (JAMA Dermatol. 2016 Apr;152[4]:452-6).

Lots more work remains to be done in order to optimize combination therapy utilizing oral itraconazole for advanced BCC. Different dosing regimens and combinations of hedgehog pathway inhibitors need to be studied. Another important question is how effective itraconazole-based combinations will be in vismodegib-naive as compared with vismodegib-resistant patients, Dr. Leitenberger observed.

He reported having no financial conflicts regarding his presentation.

Publications
Publications
Topics
Article Type
Sections
Article Source

EXPERT ANALYSIS FROM THE ACMS ANNUAL MEETING

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica