LayerRx Mapping ID
453
Slot System
Featured Buckets
Featured Buckets Admin
Reverse Chronological Sort
Allow Teaser Image
Medscape Lead Concept
65

Polypoid Melanoma: An Aggressive Variant of Nodular Melanoma

Article Type
Changed
Thu, 01/10/2019 - 13:51
Display Headline
Polypoid Melanoma: An Aggressive Variant of Nodular Melanoma

To the Editor:

An 81-year-old man presented with a nodular polypoid lesion that developed on a flat lesion on the back of 2 years’ duration. The lesion grew progressively over the course of 3 months prior to presentation. The patient had a history of melanoma in situ on the forehead that was treated with conventional surgery with clear surgical margins 6 years prior to the current presentation.

On physical examination the patient had a 4×2-cm ulcerated polypoid lesion on the back. The lesion was pink with a pigmented base. Additionally, 2 pink papules with superficial telangiectases were observed around the main lesion (Figure 1).

The gross section showed an exophytic tumor largely growing above the skin surface (Figure 2). Histopathologic analysis revealed an ulcerated lesion consisting of confluent nest and sheets of epithelioid and spindle atypical cells with numerous mitotic figures and necrotic foci (Figure 3). The thickness of the lesion was 2200 µm, and the mitotic count was 28 mitoses/mm2. There also was peritumoral vascular invasion and satellite metastasis within the perilesional hypodermis measuring 0.4 mm. Immunohistochemistry staining for S-100, human melanoma black 45 (HMB-45)(Figure 4), and Melan-A was positive in neoplastic cells.

Figure1
Figure 1. Polypoid melanoma with a 4 × 2-cm ulcerated nodular lesion with crusts on the surface and 2 satellite pink lesions around the main lesion.

Figure2
Figure 2. A longitudinal section of the polypoid tumor.

Figure3
Figure 3. Histopathology showed neoplastic proliferation with atypical epithelioid and spindle-shaped melanocytes arranged in nests with superficial ulcer (H&E, original magnification ×100).

Figure4
Figure 4. Tumor cells were diffusely positive with human melanoma black 45 (HMB-45) immunostain (original magnification ×200).

The dissemination study revealed multiple mediastinal and axillary lymphadenopathies and lesions with metastatic appearance in the brain, liver, pancreas, and muscle, together with peritoneal carcinomatosis. The patient was lost to follow-up and did not follow coadjuvant therapy with interferon alfa.

Polypoid melanoma initially was described as a type of melanoma characterized by an exophytic growth in which most of the tumor is located on the cutaneous surface, together with ulceration.1 It usually occurs in patients aged 20 to 39 years,2 and the reported incidence ranges from 1.9% to 43.3%.1 It more commonly affects mucosae, including the upper respiratory tract, esophagus, and vagina. Polypoid melanoma has a rapid progression and a poor prognosis.3 Polypoid melanoma involving the skin primarily affects the back and has a 5-year survival rate of 32% to 42%.4 Poor prognosis has been attributed to the high risk for vascular embolization under the lesion.5 Histologically, there is marked cell atypia with nuclear and cellular pleomorphism and a high mitotic count. The tumor rarely involves the reticular dermis.1,2

Polypoid melanomas are rare; however, reported frequency rates cover a wide range. These frequency rates may be due to the definition of polypoid melanoma used by the pathologist issuing the report. One of the most accepted definitions at present is a pigmented macule that progresses in months with a rapid vertical growth, invading the epidermis and the papillary dermis.2 The differential diagnosis includes pyogenic granuloma, squamous cell carcinoma, basal cell carcinoma, soft tissue sarcomas, and hemangioma.

Although our patient had a history of melanoma and the polypoid lesion developed from a flat lesion, he was late to seek medical care. The diagnosis of melanoma is made on increasingly smaller lesions with better prognosis, but there still are reports of larger melanomas. This case highlights the role dermatologists serve in the education of patients on their diagnoses and risk factors so that we may be able to diagnose non–life-threatening small lesions. It is important to remember this morphologic variety of melanoma and highlight its rapid progression and poor prognosis.

References
  1. Knezević F, Duancić V, Sitić S, et al. Histological types of polypoid cutaneous melanoma II. Coll Antropol. 2007;31:1049-1053.
  2. Dini M, Quercioli F, Caldarella V, et al. Head and neck polypoid melanoma. J Craniofac Surg. 2012;23:E23-E25.
  3. Plotnick H, Rachmaninoff N, VandenBerg HJ Jr. Polypoid melanoma: a virulent variant of nodular melanoma. report of three cases and literature review. J Am Acad Dermatol. 1990;23(5, pt 1):880-884.
  4. Manci EA, Balch CM, Murad TM, et al. Polypoid melanoma, a virulent variant of the nodular growth pattern. Am J Clin Pathol. 1981;75:810-815.
  5. De Giorgi V, Massi D, Gerlini G, et al. Immediate local and regional recurrence after the excision of a polypoid melanoma: tumor dormancy or tumor activation? Dermatol Surg. 2003;29:664-667.
Article PDF
Author and Disclosure Information

Drs. Pérez-Wilson and Castro are from Clínica Alemana de Santiago, Chile. Dr. Pérez-Wilson is from the Dermatology Department and Dr. Castro is from the Pathology Department. Dr. Arellano is from the Department of Dermatology, Universidad de Chile, Santiago.

The authors report no conflict of interest.

Correspondence: Jaime Pérez-Wilson, MD, Vitacura 5951, Santiago, Chile (jjperezw@yahoo.es).

Issue
Cutis - 101(6)
Publications
Topics
Page Number
E6-E7
Sections
Author and Disclosure Information

Drs. Pérez-Wilson and Castro are from Clínica Alemana de Santiago, Chile. Dr. Pérez-Wilson is from the Dermatology Department and Dr. Castro is from the Pathology Department. Dr. Arellano is from the Department of Dermatology, Universidad de Chile, Santiago.

The authors report no conflict of interest.

Correspondence: Jaime Pérez-Wilson, MD, Vitacura 5951, Santiago, Chile (jjperezw@yahoo.es).

Author and Disclosure Information

Drs. Pérez-Wilson and Castro are from Clínica Alemana de Santiago, Chile. Dr. Pérez-Wilson is from the Dermatology Department and Dr. Castro is from the Pathology Department. Dr. Arellano is from the Department of Dermatology, Universidad de Chile, Santiago.

The authors report no conflict of interest.

Correspondence: Jaime Pérez-Wilson, MD, Vitacura 5951, Santiago, Chile (jjperezw@yahoo.es).

Article PDF
Article PDF

To the Editor:

An 81-year-old man presented with a nodular polypoid lesion that developed on a flat lesion on the back of 2 years’ duration. The lesion grew progressively over the course of 3 months prior to presentation. The patient had a history of melanoma in situ on the forehead that was treated with conventional surgery with clear surgical margins 6 years prior to the current presentation.

On physical examination the patient had a 4×2-cm ulcerated polypoid lesion on the back. The lesion was pink with a pigmented base. Additionally, 2 pink papules with superficial telangiectases were observed around the main lesion (Figure 1).

The gross section showed an exophytic tumor largely growing above the skin surface (Figure 2). Histopathologic analysis revealed an ulcerated lesion consisting of confluent nest and sheets of epithelioid and spindle atypical cells with numerous mitotic figures and necrotic foci (Figure 3). The thickness of the lesion was 2200 µm, and the mitotic count was 28 mitoses/mm2. There also was peritumoral vascular invasion and satellite metastasis within the perilesional hypodermis measuring 0.4 mm. Immunohistochemistry staining for S-100, human melanoma black 45 (HMB-45)(Figure 4), and Melan-A was positive in neoplastic cells.

Figure1
Figure 1. Polypoid melanoma with a 4 × 2-cm ulcerated nodular lesion with crusts on the surface and 2 satellite pink lesions around the main lesion.

Figure2
Figure 2. A longitudinal section of the polypoid tumor.

Figure3
Figure 3. Histopathology showed neoplastic proliferation with atypical epithelioid and spindle-shaped melanocytes arranged in nests with superficial ulcer (H&E, original magnification ×100).

Figure4
Figure 4. Tumor cells were diffusely positive with human melanoma black 45 (HMB-45) immunostain (original magnification ×200).

The dissemination study revealed multiple mediastinal and axillary lymphadenopathies and lesions with metastatic appearance in the brain, liver, pancreas, and muscle, together with peritoneal carcinomatosis. The patient was lost to follow-up and did not follow coadjuvant therapy with interferon alfa.

Polypoid melanoma initially was described as a type of melanoma characterized by an exophytic growth in which most of the tumor is located on the cutaneous surface, together with ulceration.1 It usually occurs in patients aged 20 to 39 years,2 and the reported incidence ranges from 1.9% to 43.3%.1 It more commonly affects mucosae, including the upper respiratory tract, esophagus, and vagina. Polypoid melanoma has a rapid progression and a poor prognosis.3 Polypoid melanoma involving the skin primarily affects the back and has a 5-year survival rate of 32% to 42%.4 Poor prognosis has been attributed to the high risk for vascular embolization under the lesion.5 Histologically, there is marked cell atypia with nuclear and cellular pleomorphism and a high mitotic count. The tumor rarely involves the reticular dermis.1,2

Polypoid melanomas are rare; however, reported frequency rates cover a wide range. These frequency rates may be due to the definition of polypoid melanoma used by the pathologist issuing the report. One of the most accepted definitions at present is a pigmented macule that progresses in months with a rapid vertical growth, invading the epidermis and the papillary dermis.2 The differential diagnosis includes pyogenic granuloma, squamous cell carcinoma, basal cell carcinoma, soft tissue sarcomas, and hemangioma.

Although our patient had a history of melanoma and the polypoid lesion developed from a flat lesion, he was late to seek medical care. The diagnosis of melanoma is made on increasingly smaller lesions with better prognosis, but there still are reports of larger melanomas. This case highlights the role dermatologists serve in the education of patients on their diagnoses and risk factors so that we may be able to diagnose non–life-threatening small lesions. It is important to remember this morphologic variety of melanoma and highlight its rapid progression and poor prognosis.

To the Editor:

An 81-year-old man presented with a nodular polypoid lesion that developed on a flat lesion on the back of 2 years’ duration. The lesion grew progressively over the course of 3 months prior to presentation. The patient had a history of melanoma in situ on the forehead that was treated with conventional surgery with clear surgical margins 6 years prior to the current presentation.

On physical examination the patient had a 4×2-cm ulcerated polypoid lesion on the back. The lesion was pink with a pigmented base. Additionally, 2 pink papules with superficial telangiectases were observed around the main lesion (Figure 1).

The gross section showed an exophytic tumor largely growing above the skin surface (Figure 2). Histopathologic analysis revealed an ulcerated lesion consisting of confluent nest and sheets of epithelioid and spindle atypical cells with numerous mitotic figures and necrotic foci (Figure 3). The thickness of the lesion was 2200 µm, and the mitotic count was 28 mitoses/mm2. There also was peritumoral vascular invasion and satellite metastasis within the perilesional hypodermis measuring 0.4 mm. Immunohistochemistry staining for S-100, human melanoma black 45 (HMB-45)(Figure 4), and Melan-A was positive in neoplastic cells.

Figure1
Figure 1. Polypoid melanoma with a 4 × 2-cm ulcerated nodular lesion with crusts on the surface and 2 satellite pink lesions around the main lesion.

Figure2
Figure 2. A longitudinal section of the polypoid tumor.

Figure3
Figure 3. Histopathology showed neoplastic proliferation with atypical epithelioid and spindle-shaped melanocytes arranged in nests with superficial ulcer (H&E, original magnification ×100).

Figure4
Figure 4. Tumor cells were diffusely positive with human melanoma black 45 (HMB-45) immunostain (original magnification ×200).

The dissemination study revealed multiple mediastinal and axillary lymphadenopathies and lesions with metastatic appearance in the brain, liver, pancreas, and muscle, together with peritoneal carcinomatosis. The patient was lost to follow-up and did not follow coadjuvant therapy with interferon alfa.

Polypoid melanoma initially was described as a type of melanoma characterized by an exophytic growth in which most of the tumor is located on the cutaneous surface, together with ulceration.1 It usually occurs in patients aged 20 to 39 years,2 and the reported incidence ranges from 1.9% to 43.3%.1 It more commonly affects mucosae, including the upper respiratory tract, esophagus, and vagina. Polypoid melanoma has a rapid progression and a poor prognosis.3 Polypoid melanoma involving the skin primarily affects the back and has a 5-year survival rate of 32% to 42%.4 Poor prognosis has been attributed to the high risk for vascular embolization under the lesion.5 Histologically, there is marked cell atypia with nuclear and cellular pleomorphism and a high mitotic count. The tumor rarely involves the reticular dermis.1,2

Polypoid melanomas are rare; however, reported frequency rates cover a wide range. These frequency rates may be due to the definition of polypoid melanoma used by the pathologist issuing the report. One of the most accepted definitions at present is a pigmented macule that progresses in months with a rapid vertical growth, invading the epidermis and the papillary dermis.2 The differential diagnosis includes pyogenic granuloma, squamous cell carcinoma, basal cell carcinoma, soft tissue sarcomas, and hemangioma.

Although our patient had a history of melanoma and the polypoid lesion developed from a flat lesion, he was late to seek medical care. The diagnosis of melanoma is made on increasingly smaller lesions with better prognosis, but there still are reports of larger melanomas. This case highlights the role dermatologists serve in the education of patients on their diagnoses and risk factors so that we may be able to diagnose non–life-threatening small lesions. It is important to remember this morphologic variety of melanoma and highlight its rapid progression and poor prognosis.

References
  1. Knezević F, Duancić V, Sitić S, et al. Histological types of polypoid cutaneous melanoma II. Coll Antropol. 2007;31:1049-1053.
  2. Dini M, Quercioli F, Caldarella V, et al. Head and neck polypoid melanoma. J Craniofac Surg. 2012;23:E23-E25.
  3. Plotnick H, Rachmaninoff N, VandenBerg HJ Jr. Polypoid melanoma: a virulent variant of nodular melanoma. report of three cases and literature review. J Am Acad Dermatol. 1990;23(5, pt 1):880-884.
  4. Manci EA, Balch CM, Murad TM, et al. Polypoid melanoma, a virulent variant of the nodular growth pattern. Am J Clin Pathol. 1981;75:810-815.
  5. De Giorgi V, Massi D, Gerlini G, et al. Immediate local and regional recurrence after the excision of a polypoid melanoma: tumor dormancy or tumor activation? Dermatol Surg. 2003;29:664-667.
References
  1. Knezević F, Duancić V, Sitić S, et al. Histological types of polypoid cutaneous melanoma II. Coll Antropol. 2007;31:1049-1053.
  2. Dini M, Quercioli F, Caldarella V, et al. Head and neck polypoid melanoma. J Craniofac Surg. 2012;23:E23-E25.
  3. Plotnick H, Rachmaninoff N, VandenBerg HJ Jr. Polypoid melanoma: a virulent variant of nodular melanoma. report of three cases and literature review. J Am Acad Dermatol. 1990;23(5, pt 1):880-884.
  4. Manci EA, Balch CM, Murad TM, et al. Polypoid melanoma, a virulent variant of the nodular growth pattern. Am J Clin Pathol. 1981;75:810-815.
  5. De Giorgi V, Massi D, Gerlini G, et al. Immediate local and regional recurrence after the excision of a polypoid melanoma: tumor dormancy or tumor activation? Dermatol Surg. 2003;29:664-667.
Issue
Cutis - 101(6)
Issue
Cutis - 101(6)
Page Number
E6-E7
Page Number
E6-E7
Publications
Publications
Topics
Article Type
Display Headline
Polypoid Melanoma: An Aggressive Variant of Nodular Melanoma
Display Headline
Polypoid Melanoma: An Aggressive Variant of Nodular Melanoma
Sections
Inside the Article

Practice Points

  • The differential diagnosis of polypoid melanoma includes pyogenic granuloma and squamous cell carcinoma.
  • Polypoid melanoma has a poor prognosis because of its thickness and ulceration at the time of diagnosis and the risk of vascular embolization.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Article PDF Media

IMPACT study: Matched targeted therapy improves survival in advanced cancer

Article Type
Changed
Wed, 01/04/2023 - 16:45

 

Matched targeted therapy improved long-term survival in patients with advanced cancer, according to findings from a retrospective analysis of molecularly profiled patients.

Of 3,743 patients tested as part of IMPACT (Initiative for Molecular Profiling and Advanced Cancer Therapy), 1,307 (34.9%) had at least one targetable molecular alteration. Of those, 711 (54.4%) received either matched targeted therapy that was being tested in a clinical trial or – in a small number of cases – therapy with an approved treatment used off label, and 596 (45.6%) received nonmatched therapy, Apostolia-Maria Tsimberidou, MD, reported during a press briefing at the annual meeting of the American Society of Clinical Oncology.

The objective response rates in 697 evaluable matched therapy patients was 16.2% versus 5.4% in 571 evaluable nonmatched patients, and stable disease for at least 6 months occurred in 18.7% and 14.7% of patients, respectively, for an overall disease control rate of 34.9% versus 20.1%, said Dr. Tsimberidou, a professor at the University of Texas MD Anderson Cancer Center, Houston.

Median progression-free survival in those who received matched versus nonmatched therapy was 4.0 months and 2.8 months, respectively (hazard ratio, 0.67), and median overall survival was 9.3 and 7.3 months, respectively (HR, 0.72), she said.

The 3-year overall survival rate was 15% versus 7%, respectively, and 10-year survival was 6% and 1%, respectively.

Patients included in IMPACT had a mean age of 57 years, and 39% were men. They were heavily pretreated (mean number of prior therapies was 4); only 2.8% of patients had no prior treatment. Cancers included gastrointestinal (24.2%), gynecologic (19.4%), breast (13.5%), melanoma (11.9%) and lung (8.7%).

In this video interview, Dr. Tsimberidou describes the rationale, methodology, and findings of IMPACT, including the use of a prognostic scoring system developed as part of the study to predict overall survival based on baseline characteristics, such as baseline p13K/AKT/mTOR pathway molecular alterations, which were shown on multivariate analysis in IMPACT to predict shorter overall survival versus other alterations. Other predictors of shorter survival included liver metastases, elevated lactate dehydrogenase levels, poor functional status, low albumin levels, elevated platelet counts, and age of 60 years or older.

 

 


“We [also] wanted to see if adding the intervention ... would hold significance in this multivariate model, and we found that ... nonmatched therapy was associated with adverse survival; it was an independent factor associated with worse survival,” she said. “Therefore, matched targeted therapy is associated with longer survival.”

In the randomized, phase 2 trial IMPACT 2, progression-free survival will be compared in patients with and without matched targeted therapy, and the prognostic scoring system developed as part of IMPACT to predict overall survival based on baseline characteristics will be further evaluated, she said.

During a discussion of the findings during the press briefing, ASCO Expert Catherine M. Diefenbach, MD, said the type of precision medicine studied in IMPACT is “the wave of the future.

“Large scale efforts such as ASCO’s TAPUR or the NCI-MATCH trial will bring these efforts to many, many more patients, and hopefully usher in a new way of treating advanced cancer patients that will improve overall survival for many more patients,” said Dr. Diefenbach, of New York University.

Dr. Tsimberidou reported a consulting or advisory role with Roche, as well as research funding to her institution from EMD Serono, Baxter, Foundation Medicine, ONYX, Bayer, Boston Biomedical, Placon, IMMATICS, Karus Therapeutics, and StemCells.

SOURCE: Tsimberidou AM et al. ASCO 2018, Abstract LBA 2553.

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

 

Matched targeted therapy improved long-term survival in patients with advanced cancer, according to findings from a retrospective analysis of molecularly profiled patients.

Of 3,743 patients tested as part of IMPACT (Initiative for Molecular Profiling and Advanced Cancer Therapy), 1,307 (34.9%) had at least one targetable molecular alteration. Of those, 711 (54.4%) received either matched targeted therapy that was being tested in a clinical trial or – in a small number of cases – therapy with an approved treatment used off label, and 596 (45.6%) received nonmatched therapy, Apostolia-Maria Tsimberidou, MD, reported during a press briefing at the annual meeting of the American Society of Clinical Oncology.

The objective response rates in 697 evaluable matched therapy patients was 16.2% versus 5.4% in 571 evaluable nonmatched patients, and stable disease for at least 6 months occurred in 18.7% and 14.7% of patients, respectively, for an overall disease control rate of 34.9% versus 20.1%, said Dr. Tsimberidou, a professor at the University of Texas MD Anderson Cancer Center, Houston.

Median progression-free survival in those who received matched versus nonmatched therapy was 4.0 months and 2.8 months, respectively (hazard ratio, 0.67), and median overall survival was 9.3 and 7.3 months, respectively (HR, 0.72), she said.

The 3-year overall survival rate was 15% versus 7%, respectively, and 10-year survival was 6% and 1%, respectively.

Patients included in IMPACT had a mean age of 57 years, and 39% were men. They were heavily pretreated (mean number of prior therapies was 4); only 2.8% of patients had no prior treatment. Cancers included gastrointestinal (24.2%), gynecologic (19.4%), breast (13.5%), melanoma (11.9%) and lung (8.7%).

In this video interview, Dr. Tsimberidou describes the rationale, methodology, and findings of IMPACT, including the use of a prognostic scoring system developed as part of the study to predict overall survival based on baseline characteristics, such as baseline p13K/AKT/mTOR pathway molecular alterations, which were shown on multivariate analysis in IMPACT to predict shorter overall survival versus other alterations. Other predictors of shorter survival included liver metastases, elevated lactate dehydrogenase levels, poor functional status, low albumin levels, elevated platelet counts, and age of 60 years or older.

 

 


“We [also] wanted to see if adding the intervention ... would hold significance in this multivariate model, and we found that ... nonmatched therapy was associated with adverse survival; it was an independent factor associated with worse survival,” she said. “Therefore, matched targeted therapy is associated with longer survival.”

In the randomized, phase 2 trial IMPACT 2, progression-free survival will be compared in patients with and without matched targeted therapy, and the prognostic scoring system developed as part of IMPACT to predict overall survival based on baseline characteristics will be further evaluated, she said.

During a discussion of the findings during the press briefing, ASCO Expert Catherine M. Diefenbach, MD, said the type of precision medicine studied in IMPACT is “the wave of the future.

“Large scale efforts such as ASCO’s TAPUR or the NCI-MATCH trial will bring these efforts to many, many more patients, and hopefully usher in a new way of treating advanced cancer patients that will improve overall survival for many more patients,” said Dr. Diefenbach, of New York University.

Dr. Tsimberidou reported a consulting or advisory role with Roche, as well as research funding to her institution from EMD Serono, Baxter, Foundation Medicine, ONYX, Bayer, Boston Biomedical, Placon, IMMATICS, Karus Therapeutics, and StemCells.

SOURCE: Tsimberidou AM et al. ASCO 2018, Abstract LBA 2553.

 

Matched targeted therapy improved long-term survival in patients with advanced cancer, according to findings from a retrospective analysis of molecularly profiled patients.

Of 3,743 patients tested as part of IMPACT (Initiative for Molecular Profiling and Advanced Cancer Therapy), 1,307 (34.9%) had at least one targetable molecular alteration. Of those, 711 (54.4%) received either matched targeted therapy that was being tested in a clinical trial or – in a small number of cases – therapy with an approved treatment used off label, and 596 (45.6%) received nonmatched therapy, Apostolia-Maria Tsimberidou, MD, reported during a press briefing at the annual meeting of the American Society of Clinical Oncology.

The objective response rates in 697 evaluable matched therapy patients was 16.2% versus 5.4% in 571 evaluable nonmatched patients, and stable disease for at least 6 months occurred in 18.7% and 14.7% of patients, respectively, for an overall disease control rate of 34.9% versus 20.1%, said Dr. Tsimberidou, a professor at the University of Texas MD Anderson Cancer Center, Houston.

Median progression-free survival in those who received matched versus nonmatched therapy was 4.0 months and 2.8 months, respectively (hazard ratio, 0.67), and median overall survival was 9.3 and 7.3 months, respectively (HR, 0.72), she said.

The 3-year overall survival rate was 15% versus 7%, respectively, and 10-year survival was 6% and 1%, respectively.

Patients included in IMPACT had a mean age of 57 years, and 39% were men. They were heavily pretreated (mean number of prior therapies was 4); only 2.8% of patients had no prior treatment. Cancers included gastrointestinal (24.2%), gynecologic (19.4%), breast (13.5%), melanoma (11.9%) and lung (8.7%).

In this video interview, Dr. Tsimberidou describes the rationale, methodology, and findings of IMPACT, including the use of a prognostic scoring system developed as part of the study to predict overall survival based on baseline characteristics, such as baseline p13K/AKT/mTOR pathway molecular alterations, which were shown on multivariate analysis in IMPACT to predict shorter overall survival versus other alterations. Other predictors of shorter survival included liver metastases, elevated lactate dehydrogenase levels, poor functional status, low albumin levels, elevated platelet counts, and age of 60 years or older.

 

 


“We [also] wanted to see if adding the intervention ... would hold significance in this multivariate model, and we found that ... nonmatched therapy was associated with adverse survival; it was an independent factor associated with worse survival,” she said. “Therefore, matched targeted therapy is associated with longer survival.”

In the randomized, phase 2 trial IMPACT 2, progression-free survival will be compared in patients with and without matched targeted therapy, and the prognostic scoring system developed as part of IMPACT to predict overall survival based on baseline characteristics will be further evaluated, she said.

During a discussion of the findings during the press briefing, ASCO Expert Catherine M. Diefenbach, MD, said the type of precision medicine studied in IMPACT is “the wave of the future.

“Large scale efforts such as ASCO’s TAPUR or the NCI-MATCH trial will bring these efforts to many, many more patients, and hopefully usher in a new way of treating advanced cancer patients that will improve overall survival for many more patients,” said Dr. Diefenbach, of New York University.

Dr. Tsimberidou reported a consulting or advisory role with Roche, as well as research funding to her institution from EMD Serono, Baxter, Foundation Medicine, ONYX, Bayer, Boston Biomedical, Placon, IMMATICS, Karus Therapeutics, and StemCells.

SOURCE: Tsimberidou AM et al. ASCO 2018, Abstract LBA 2553.

Publications
Publications
Topics
Article Type
Sections
Article Source

REPORTING FROM ASCO 2018

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

 

Key clinical point: Matched targeted therapy improved survival in patients with advanced cancer.

Major finding: The 3-yearoverall survival rate with matched versus nonmatched therapy was 15% and 7%, respectively.

Study details: A retrospective analysis (IMPACT) of 3,743 molecularly profiled advanced cancer patients.

Disclosures: Dr. Tsimberidou reported a consulting or advisory role with Roche, as well as research funding to her institution from EMD Serono, Baxter, Foundation Medicine, ONYX Medical, Bayer, Boston Biomedical, Placon, IMMATICS, Karus Therapeutics, and StemCells.

Source: Tsimberidou AM et al. ASCO 2018, Abstract LBA 2553.

Disqus Comments
Default
Use ProPublica

New criteria slightly improved melanoma staging

Eighth edition yields “modest” progress
Article Type
Changed
Mon, 01/14/2019 - 10:24

 

Use of the recently updated AJCC Cancer Staging Manual, 8th edition, produced about 10% better accuracy when interpreting T1a invasive melanomas in a national study.

For stage T1b or greater cases, using the AJCC 8 instead of the AJCC 7 yielded about 6% higher concordance between interpretations and consensus reference diagnoses, said Joann G. Elmore, MD, MPH, of the David Geffen School of Medicine at the University of California, Los Angeles, and her associates. “Intraobserver reproducibility of diagnoses also improved,” but “concordance and reproducibility remain low,” the researchers wrote. The report was published in JAMA Open Network.

For the study, 187 pathologists each interpreted one of five glass slide sets of melanocytic skin biopsies. At least 8 months later, they interpreted the same set again, both times recording their interpretations on the online Pathology Assessment Tool and Hierarchy for Diagnosis (MPATH-Dx). Researchers then used AJCC 7 and AJCC 8 criteria to group these interpretations into one of five MPATH-Dx classes. The final analysis included only the 116 cases of invasive melanoma, because changes in the AJCC 8 criteria affected only MPATH-Dx classes IV (T1a) and V (T1b and higher), the investigators explained.

For T1a invasive melanomas, overall concordance between interpretations and consensus diagnoses rose from 44% (95% confidence interval, 41%-48%) under the AJCC 7 criteria to 54% (95% CI, 51%-57%) under the AJCC 8 criteria. For stage T1b or higher lesions, concordance rose from 72% to 78%. Intrapathologist reproducibility improved from 59% to 64% for T1a cases and from 74% to 77% for T1b or higher cases. Thus, the AJCC 8 produced “modest” improvements in concordance and reproducibility over the AJCC 7, the researchers said.

One of the most likely explanations is that the AJCC 8 eliminated dermal mitotic activity as a criterion for T1b melanomas, which is “potentially unreliable” in thin lesions, they added. Persistently low concordance and reproducibility “suggest that conventional histopathology has been parsed to a degree that falls below the limits of reliability,” they concluded.

The National Cancer Institute provided funding. Dr. Elmore disclosed ties to Myriad Genetics, SciBase, and the National Institutes of Health.

SOURCE: Elmore JG et al. JAMA Network Open. 2018 May 18. doi: 10.1001/jamanetworkopen.2018.0083.

Body

 

Compared with the seventh edition, use of the eighth AJCC Cancer Staging Manual (AJCC 8) produced “modest but significant improvements” in reproducibility and concordance when microstaging cutaneous melanoma, Peter M. Ferguson, MBChB, PhD, FRCPA, Jeffrey E. Gershenwald, MD, and Richard A. Scolyer, MD, FRCPA, wrote in an editorial accompanying the study.

But the study revealed “significant” shortcomings in reproducibility, they added. Future studies should explore the reproducibility of individual AJCC parameters and how experience and access to relevant case data affects reproducibility, they wrote.

The study used the Melanocytic Pathology Assessment Tool and Hierarchy for Diagnosis system instead of classifying cases by AJCC tumor categories. Consequently, pathologists collapsed several AJCC tumor subcategories into the Melanoma Pathology Study class V, which limited interpretability of results for T1b melanomas, an important threshold for recommending sentinel node biopsy. “[We] have published higher rates of concordance using AJCC 7 criteria, albeit with access to clinical information and reporting by a small number of pathologists with expertise reporting melanomas at a high-volume referral center,” the editorialists emphasized.

Dr. Ferguson and Dr. Scolyer are with the University of Sydney. Dr. Gershenwald is with the University of Texas MD Anderson Cancer Center, Houston. The editorialists disclosed support from the Robert and Lynne Grossman Family Foundation, the Michael and Patricia Booker Melanoma Research Endowment, Melanoma Institute Australia, the Deborah and John McMurtrie Melanoma Institute Australia Pathology Fellowship, and the Australian National Health and Medical Research Council Fellowship. Dr. Gershenwald disclosed ties to Merck, Syndax, Castle Biosciences, and the AJCC (JAMA Network Open. 2018 May 18. doi: 10.1001/jamanetworkopen.2018.0086 ).

Publications
Topics
Sections
Body

 

Compared with the seventh edition, use of the eighth AJCC Cancer Staging Manual (AJCC 8) produced “modest but significant improvements” in reproducibility and concordance when microstaging cutaneous melanoma, Peter M. Ferguson, MBChB, PhD, FRCPA, Jeffrey E. Gershenwald, MD, and Richard A. Scolyer, MD, FRCPA, wrote in an editorial accompanying the study.

But the study revealed “significant” shortcomings in reproducibility, they added. Future studies should explore the reproducibility of individual AJCC parameters and how experience and access to relevant case data affects reproducibility, they wrote.

The study used the Melanocytic Pathology Assessment Tool and Hierarchy for Diagnosis system instead of classifying cases by AJCC tumor categories. Consequently, pathologists collapsed several AJCC tumor subcategories into the Melanoma Pathology Study class V, which limited interpretability of results for T1b melanomas, an important threshold for recommending sentinel node biopsy. “[We] have published higher rates of concordance using AJCC 7 criteria, albeit with access to clinical information and reporting by a small number of pathologists with expertise reporting melanomas at a high-volume referral center,” the editorialists emphasized.

Dr. Ferguson and Dr. Scolyer are with the University of Sydney. Dr. Gershenwald is with the University of Texas MD Anderson Cancer Center, Houston. The editorialists disclosed support from the Robert and Lynne Grossman Family Foundation, the Michael and Patricia Booker Melanoma Research Endowment, Melanoma Institute Australia, the Deborah and John McMurtrie Melanoma Institute Australia Pathology Fellowship, and the Australian National Health and Medical Research Council Fellowship. Dr. Gershenwald disclosed ties to Merck, Syndax, Castle Biosciences, and the AJCC (JAMA Network Open. 2018 May 18. doi: 10.1001/jamanetworkopen.2018.0086 ).

Body

 

Compared with the seventh edition, use of the eighth AJCC Cancer Staging Manual (AJCC 8) produced “modest but significant improvements” in reproducibility and concordance when microstaging cutaneous melanoma, Peter M. Ferguson, MBChB, PhD, FRCPA, Jeffrey E. Gershenwald, MD, and Richard A. Scolyer, MD, FRCPA, wrote in an editorial accompanying the study.

But the study revealed “significant” shortcomings in reproducibility, they added. Future studies should explore the reproducibility of individual AJCC parameters and how experience and access to relevant case data affects reproducibility, they wrote.

The study used the Melanocytic Pathology Assessment Tool and Hierarchy for Diagnosis system instead of classifying cases by AJCC tumor categories. Consequently, pathologists collapsed several AJCC tumor subcategories into the Melanoma Pathology Study class V, which limited interpretability of results for T1b melanomas, an important threshold for recommending sentinel node biopsy. “[We] have published higher rates of concordance using AJCC 7 criteria, albeit with access to clinical information and reporting by a small number of pathologists with expertise reporting melanomas at a high-volume referral center,” the editorialists emphasized.

Dr. Ferguson and Dr. Scolyer are with the University of Sydney. Dr. Gershenwald is with the University of Texas MD Anderson Cancer Center, Houston. The editorialists disclosed support from the Robert and Lynne Grossman Family Foundation, the Michael and Patricia Booker Melanoma Research Endowment, Melanoma Institute Australia, the Deborah and John McMurtrie Melanoma Institute Australia Pathology Fellowship, and the Australian National Health and Medical Research Council Fellowship. Dr. Gershenwald disclosed ties to Merck, Syndax, Castle Biosciences, and the AJCC (JAMA Network Open. 2018 May 18. doi: 10.1001/jamanetworkopen.2018.0086 ).

Title
Eighth edition yields “modest” progress
Eighth edition yields “modest” progress

 

Use of the recently updated AJCC Cancer Staging Manual, 8th edition, produced about 10% better accuracy when interpreting T1a invasive melanomas in a national study.

For stage T1b or greater cases, using the AJCC 8 instead of the AJCC 7 yielded about 6% higher concordance between interpretations and consensus reference diagnoses, said Joann G. Elmore, MD, MPH, of the David Geffen School of Medicine at the University of California, Los Angeles, and her associates. “Intraobserver reproducibility of diagnoses also improved,” but “concordance and reproducibility remain low,” the researchers wrote. The report was published in JAMA Open Network.

For the study, 187 pathologists each interpreted one of five glass slide sets of melanocytic skin biopsies. At least 8 months later, they interpreted the same set again, both times recording their interpretations on the online Pathology Assessment Tool and Hierarchy for Diagnosis (MPATH-Dx). Researchers then used AJCC 7 and AJCC 8 criteria to group these interpretations into one of five MPATH-Dx classes. The final analysis included only the 116 cases of invasive melanoma, because changes in the AJCC 8 criteria affected only MPATH-Dx classes IV (T1a) and V (T1b and higher), the investigators explained.

For T1a invasive melanomas, overall concordance between interpretations and consensus diagnoses rose from 44% (95% confidence interval, 41%-48%) under the AJCC 7 criteria to 54% (95% CI, 51%-57%) under the AJCC 8 criteria. For stage T1b or higher lesions, concordance rose from 72% to 78%. Intrapathologist reproducibility improved from 59% to 64% for T1a cases and from 74% to 77% for T1b or higher cases. Thus, the AJCC 8 produced “modest” improvements in concordance and reproducibility over the AJCC 7, the researchers said.

One of the most likely explanations is that the AJCC 8 eliminated dermal mitotic activity as a criterion for T1b melanomas, which is “potentially unreliable” in thin lesions, they added. Persistently low concordance and reproducibility “suggest that conventional histopathology has been parsed to a degree that falls below the limits of reliability,” they concluded.

The National Cancer Institute provided funding. Dr. Elmore disclosed ties to Myriad Genetics, SciBase, and the National Institutes of Health.

SOURCE: Elmore JG et al. JAMA Network Open. 2018 May 18. doi: 10.1001/jamanetworkopen.2018.0083.

 

Use of the recently updated AJCC Cancer Staging Manual, 8th edition, produced about 10% better accuracy when interpreting T1a invasive melanomas in a national study.

For stage T1b or greater cases, using the AJCC 8 instead of the AJCC 7 yielded about 6% higher concordance between interpretations and consensus reference diagnoses, said Joann G. Elmore, MD, MPH, of the David Geffen School of Medicine at the University of California, Los Angeles, and her associates. “Intraobserver reproducibility of diagnoses also improved,” but “concordance and reproducibility remain low,” the researchers wrote. The report was published in JAMA Open Network.

For the study, 187 pathologists each interpreted one of five glass slide sets of melanocytic skin biopsies. At least 8 months later, they interpreted the same set again, both times recording their interpretations on the online Pathology Assessment Tool and Hierarchy for Diagnosis (MPATH-Dx). Researchers then used AJCC 7 and AJCC 8 criteria to group these interpretations into one of five MPATH-Dx classes. The final analysis included only the 116 cases of invasive melanoma, because changes in the AJCC 8 criteria affected only MPATH-Dx classes IV (T1a) and V (T1b and higher), the investigators explained.

For T1a invasive melanomas, overall concordance between interpretations and consensus diagnoses rose from 44% (95% confidence interval, 41%-48%) under the AJCC 7 criteria to 54% (95% CI, 51%-57%) under the AJCC 8 criteria. For stage T1b or higher lesions, concordance rose from 72% to 78%. Intrapathologist reproducibility improved from 59% to 64% for T1a cases and from 74% to 77% for T1b or higher cases. Thus, the AJCC 8 produced “modest” improvements in concordance and reproducibility over the AJCC 7, the researchers said.

One of the most likely explanations is that the AJCC 8 eliminated dermal mitotic activity as a criterion for T1b melanomas, which is “potentially unreliable” in thin lesions, they added. Persistently low concordance and reproducibility “suggest that conventional histopathology has been parsed to a degree that falls below the limits of reliability,” they concluded.

The National Cancer Institute provided funding. Dr. Elmore disclosed ties to Myriad Genetics, SciBase, and the National Institutes of Health.

SOURCE: Elmore JG et al. JAMA Network Open. 2018 May 18. doi: 10.1001/jamanetworkopen.2018.0083.

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

FROM JAMA NETWORK OPEN

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

 

Key clinical point: Use of the recently updated AJCC Cancer Staging Manual, 8th edition, somewhat improved concordance and reproducibility for invasive melanoma, compared with the previous edition.

Major finding: For diagnoses of T1a invasive melanoma, concordance with consensus diagnoses rose from 44% with the AJCC 7 to 54% with the AJCC 8. For T1b, concordance rose from 72% to 78%.

Study details: Interpretations of 116 invasive melanomas by 187 pathologists.

Disclosures: The National Cancer Institute provided funding. Dr. Elmore disclosed ties to Myriad Genetics, SciBase, and the National Institutes of Health.

Source: Elmore JG et al. JAMA Network Open. 2018 May 18. doi: 10.1001/jamanetworkopen.2018.0083.

Disqus Comments
Default
Use ProPublica

Ropivacaine called top anesthesia for nail surgery

Article Type
Changed
Mon, 01/14/2019 - 10:24

 

– Ropivacaine has a fast onset of action, longer duration than either lidocaine or bupivacaine, and it’s the only one of the three that’s inherently vasoconstrictive. For Brienne Cressey, MD, those features make ropivacaine the local anesthetic of choice in performing nail surgery.

“Local anesthesia is really key for nail surgery. If you don’t have good anesthesia it’s not a good experience for either the surgeon or the patient,” she observed at the annual meeting of the American College of Mohs Surgery.

Bruce Jancin/MDedge News
Dr. Brienne Cressey
However, when she asked for a show of hands at the outset of her talk on nail surgery pearls before a packed hall filled with Mohs surgeons, lidocaine (Xylocaine) was far and away the audience’s number-one choice for local anesthesia in nail surgery, followed by bupivacaine (Marcaine) in a distant second. Only a handful of surgeons favored ropivacaine (Naropin). So Dr. Cressey set about convincing the crowd as to why ropivacaine is worthy of serious consideration. She also explained how she employs the local anesthetic in performing a distal digital block, a procedure not widely used in dermatology, but one she prefers for nail surgery.

Lidocaine has a fast onset – less than 1 minute – but a problematic short duration of 30-120 minutes. Bupivacaine has the disadvantage of a slow onset of up to 5 minutes, albeit with a longer duration of anesthesia at 2-4 hours. Ropivacaine has a fast onset, plus a duration of up to 8 hours. And unlike lidocaine and bupivacaine, which are vasodilatory, ropivacaine is vasoconstrictive.



“With lidocaine, you get a lot of blood right after you take off your tourniquet. With ropivacaine, you get really nice reperfusion, but it’s not too much. You take off the tourniquet, check to see you’ve got reperfusion, then you add a little ropivacaine – about 0.5 mL – on either side of the base of the distal phalanx. It stops the bleeding immediately and you can easily put on a pressure dressing. It’s a nice way to get the patient over the hump of those first hours of pain and lets them drive home in comfort,” explained Dr. Cressey, a dermatologist working in a group practice at Dermatology Professionals in East Greenwich, R.I.

Ropivacaine is less cardiotoxic than bupivacaine. And ropivacaine offers an additional advantage: Its pH is such that no buffering is necessary. “Ropivacaine doesn’t require any compounding. You can just use it at 1% straight out of the bottle. That’s what we do in our office, and we’ve had very good experience with it,” according to the dermatologist.

Achieving smooth sailing with local anesthesia

Dr. Cressey delivers ropivacaine slowly through a 30-gauge needle, which makes for a smaller, less painful puncture. She utilizes a topical cold spray, and places a vibrating machine as a distractant proximal to where she is injecting. She keeps the anesthetic at room temperature or warms it to body temperature in a water bath as another means of reducing the pain of injection.

 

 

The distal digital block

This is a cross between a traditional proximal digital block and a wing block. It works well for the second, third, and fourth digits, which are mostly volar dominant. The block bathes the volar nerve branch in anesthesia at the midline of the finger or toe.

Dr. Cressey begins by injecting ropivacaine proximal and lateral to the junction of the proximal nail fold and lateral nail fold. After creating a dermal wheal, she directs her needle perpendicularly downward toward the finger or toe pad, injecting 1-4 mL of anesthesia, depending upon digit size. Visible blanching will progress digitally. If resistance is encountered, it suggests the needle has penetrated a ligament or other fibrous tissue. Simply withdraw the needle and continue injecting.

“What’s nice about the distal digital block is you get an immediate effect, and there’s good hemostasis during the procedure as well,” she said.

Dr. Cressey reported no financial conflicts regarding her presentation.

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

 

– Ropivacaine has a fast onset of action, longer duration than either lidocaine or bupivacaine, and it’s the only one of the three that’s inherently vasoconstrictive. For Brienne Cressey, MD, those features make ropivacaine the local anesthetic of choice in performing nail surgery.

“Local anesthesia is really key for nail surgery. If you don’t have good anesthesia it’s not a good experience for either the surgeon or the patient,” she observed at the annual meeting of the American College of Mohs Surgery.

Bruce Jancin/MDedge News
Dr. Brienne Cressey
However, when she asked for a show of hands at the outset of her talk on nail surgery pearls before a packed hall filled with Mohs surgeons, lidocaine (Xylocaine) was far and away the audience’s number-one choice for local anesthesia in nail surgery, followed by bupivacaine (Marcaine) in a distant second. Only a handful of surgeons favored ropivacaine (Naropin). So Dr. Cressey set about convincing the crowd as to why ropivacaine is worthy of serious consideration. She also explained how she employs the local anesthetic in performing a distal digital block, a procedure not widely used in dermatology, but one she prefers for nail surgery.

Lidocaine has a fast onset – less than 1 minute – but a problematic short duration of 30-120 minutes. Bupivacaine has the disadvantage of a slow onset of up to 5 minutes, albeit with a longer duration of anesthesia at 2-4 hours. Ropivacaine has a fast onset, plus a duration of up to 8 hours. And unlike lidocaine and bupivacaine, which are vasodilatory, ropivacaine is vasoconstrictive.



“With lidocaine, you get a lot of blood right after you take off your tourniquet. With ropivacaine, you get really nice reperfusion, but it’s not too much. You take off the tourniquet, check to see you’ve got reperfusion, then you add a little ropivacaine – about 0.5 mL – on either side of the base of the distal phalanx. It stops the bleeding immediately and you can easily put on a pressure dressing. It’s a nice way to get the patient over the hump of those first hours of pain and lets them drive home in comfort,” explained Dr. Cressey, a dermatologist working in a group practice at Dermatology Professionals in East Greenwich, R.I.

Ropivacaine is less cardiotoxic than bupivacaine. And ropivacaine offers an additional advantage: Its pH is such that no buffering is necessary. “Ropivacaine doesn’t require any compounding. You can just use it at 1% straight out of the bottle. That’s what we do in our office, and we’ve had very good experience with it,” according to the dermatologist.

Achieving smooth sailing with local anesthesia

Dr. Cressey delivers ropivacaine slowly through a 30-gauge needle, which makes for a smaller, less painful puncture. She utilizes a topical cold spray, and places a vibrating machine as a distractant proximal to where she is injecting. She keeps the anesthetic at room temperature or warms it to body temperature in a water bath as another means of reducing the pain of injection.

 

 

The distal digital block

This is a cross between a traditional proximal digital block and a wing block. It works well for the second, third, and fourth digits, which are mostly volar dominant. The block bathes the volar nerve branch in anesthesia at the midline of the finger or toe.

Dr. Cressey begins by injecting ropivacaine proximal and lateral to the junction of the proximal nail fold and lateral nail fold. After creating a dermal wheal, she directs her needle perpendicularly downward toward the finger or toe pad, injecting 1-4 mL of anesthesia, depending upon digit size. Visible blanching will progress digitally. If resistance is encountered, it suggests the needle has penetrated a ligament or other fibrous tissue. Simply withdraw the needle and continue injecting.

“What’s nice about the distal digital block is you get an immediate effect, and there’s good hemostasis during the procedure as well,” she said.

Dr. Cressey reported no financial conflicts regarding her presentation.

 

– Ropivacaine has a fast onset of action, longer duration than either lidocaine or bupivacaine, and it’s the only one of the three that’s inherently vasoconstrictive. For Brienne Cressey, MD, those features make ropivacaine the local anesthetic of choice in performing nail surgery.

“Local anesthesia is really key for nail surgery. If you don’t have good anesthesia it’s not a good experience for either the surgeon or the patient,” she observed at the annual meeting of the American College of Mohs Surgery.

Bruce Jancin/MDedge News
Dr. Brienne Cressey
However, when she asked for a show of hands at the outset of her talk on nail surgery pearls before a packed hall filled with Mohs surgeons, lidocaine (Xylocaine) was far and away the audience’s number-one choice for local anesthesia in nail surgery, followed by bupivacaine (Marcaine) in a distant second. Only a handful of surgeons favored ropivacaine (Naropin). So Dr. Cressey set about convincing the crowd as to why ropivacaine is worthy of serious consideration. She also explained how she employs the local anesthetic in performing a distal digital block, a procedure not widely used in dermatology, but one she prefers for nail surgery.

Lidocaine has a fast onset – less than 1 minute – but a problematic short duration of 30-120 minutes. Bupivacaine has the disadvantage of a slow onset of up to 5 minutes, albeit with a longer duration of anesthesia at 2-4 hours. Ropivacaine has a fast onset, plus a duration of up to 8 hours. And unlike lidocaine and bupivacaine, which are vasodilatory, ropivacaine is vasoconstrictive.



“With lidocaine, you get a lot of blood right after you take off your tourniquet. With ropivacaine, you get really nice reperfusion, but it’s not too much. You take off the tourniquet, check to see you’ve got reperfusion, then you add a little ropivacaine – about 0.5 mL – on either side of the base of the distal phalanx. It stops the bleeding immediately and you can easily put on a pressure dressing. It’s a nice way to get the patient over the hump of those first hours of pain and lets them drive home in comfort,” explained Dr. Cressey, a dermatologist working in a group practice at Dermatology Professionals in East Greenwich, R.I.

Ropivacaine is less cardiotoxic than bupivacaine. And ropivacaine offers an additional advantage: Its pH is such that no buffering is necessary. “Ropivacaine doesn’t require any compounding. You can just use it at 1% straight out of the bottle. That’s what we do in our office, and we’ve had very good experience with it,” according to the dermatologist.

Achieving smooth sailing with local anesthesia

Dr. Cressey delivers ropivacaine slowly through a 30-gauge needle, which makes for a smaller, less painful puncture. She utilizes a topical cold spray, and places a vibrating machine as a distractant proximal to where she is injecting. She keeps the anesthetic at room temperature or warms it to body temperature in a water bath as another means of reducing the pain of injection.

 

 

The distal digital block

This is a cross between a traditional proximal digital block and a wing block. It works well for the second, third, and fourth digits, which are mostly volar dominant. The block bathes the volar nerve branch in anesthesia at the midline of the finger or toe.

Dr. Cressey begins by injecting ropivacaine proximal and lateral to the junction of the proximal nail fold and lateral nail fold. After creating a dermal wheal, she directs her needle perpendicularly downward toward the finger or toe pad, injecting 1-4 mL of anesthesia, depending upon digit size. Visible blanching will progress digitally. If resistance is encountered, it suggests the needle has penetrated a ligament or other fibrous tissue. Simply withdraw the needle and continue injecting.

“What’s nice about the distal digital block is you get an immediate effect, and there’s good hemostasis during the procedure as well,” she said.

Dr. Cressey reported no financial conflicts regarding her presentation.

Publications
Publications
Topics
Article Type
Click for Credit Status
Ready
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

Using Dermoscopy to Identify Melanoma and Improve Diagnostic Discrimination (FULL)

Article Type
Changed
Thu, 12/15/2022 - 14:51
Display Headline
Using Dermoscopy to Identify Melanoma and Improve Diagnostic Discrimination
Use of dermoscopy and detection algorithms by primary care physicians can enhance assessment of clinically suspicious lesions compared with that of naked eye examinations.

From 1982 to 2011, the melanoma incidence rate doubled in the US.1 In 2018, an estimated 87,290 cases of melanoma in situ and 91,270 cases of invasive melanoma will be diagnosed in the US, and 9,320 deaths will be attributable to melanoma.2 Early detection of melanoma is critically important to reduce melanoma-related mortality, with 5-year survival rates as high as 97% at stage 1A vs a 20% 5-year survival when there is distant metastasis.2,3 Melanoma is particularly relevant for medical providers working with veterans because melanoma disproportionately affects service members with an incidence rate ratio of 1.62 (95% confidence interval [CI], 1.40-1.86) compared with that of the general population.4

Biopsy is the definitive diagnostic tool for melanoma. Histologic analysis differentiates melanoma from seborrheic keratoses, pigmented nevi, dermatofibromas, and other pigmented lesions that can resemble melanoma on clinical examination. However, biopsy must be used judiciously as unnecessary biopsies contribute to health care costs and leave scars, which can have psychosocial implications. With benign nevi outnumbering melanoma about 2 million to 1, biopsy is indicated once a threshold of suspicion is obtained.5

Dermoscopic Tool

Dermoscopy is a microscopy-based tool to improve noninvasive diagnostic discrimination of skin lesions based on color and structure analysis. Coloration provides an indication of the composition of elements present in the skin with keratin appearing yellow, blood appearing red, and collagen appearing white. Coloration also suggests pigment depth as melanin appears black when located in the stratum corneum, brown when located deeper in the epidermis, and blue when located in the dermis.6 Finally, characteristic histopathologic alterations in the dermoepidermal junction, rete ridges, pigment-containing cells, and/or melanocyte granules that occur in melanoma are recognizable with dermoscopy.6

In 2001, Bafounta and colleagues performed the first meta-analysis on the efficacy of dermoscopy compared with that of clinical evaluation, finding that dermoscopy performed specifically by dermatology-trained clinicians improved the accuracy of identifying melanoma from an odds ratio of 16 (95% CI, 9-31) with naked eye examination to 76 (95% CI, 25-223) with dermoscopy.7

More recently, Terushkin and colleagues demonstrated that diagnosis specificity improves when a general dermatologist is trained in dermoscopic pattern recognition. Naked eye examination produced a benign to malignant ratio (BMR) of 18.4:1, indicating that about 18 of 19 biopsies considered suspicious for melanoma ultimately yielded benign melanocytic lesions. Although the BMR for the general dermatologist experienced an increase after dermoscopy training, the ratio eventually decreased to 7.9:1.8

Dermoscopic Analysis

Some of the common patterns recognized in melanoma are demonstrated in Figures 1 and 2. Figure 1 is a close-up of a patient’s upper back showing a solitary asymmetric melanocytic lesion containing multiple red, brown, black, and blue hues. 

The lesion is highly suspicious for melanoma. Key patterns identified under dermoscopy in Figure 2 increase the level of suspicion. The pink circle in the upper left of the lesion demonstrates a scarlike regression of pigment structure. 
The orange triangle signifies a region with marked variability in color called an atypical pigment network, and the centrally located yellow circle and gray square identify interspersed atypical dots and globules of color. The red rectangle on the right highlights irregular streaking, linear radial projections suggestive of superficial spreading melanoma. The green line identifies hypopigmentation with surrounding curvilinear globular structures collectively known as a negative network.  Finally, the bottom blue triangle overlies an area with a hazy blue tinge called a blue-white veil, indicating the presence of melanocytes deep in the dermis (Table 1).6,9

Pattern analysis, the dermoscopic interpretation method preferred by pigmented lesion specialists, requires simultaneously assessing numerous lesion patterns that vary depending on body site.10 Alternative dermoscopic algorithms that focus on the most common features of melanoma have been developed to aid practitioners with the interpretation of dermoscopy findings: the 7-point checklist, the Menzies method, the ABCD rule, and the CASH algorithm (Tables 2, 3, 4, and 5). 

To apply these algorithms to evaluate the lesion in Figures 1 and 2 ( eAppendix ).11-14 The triage amalgamated dermoscopic algorithm (TADA) method, a newer algorithm designed for novice dermoscopy users, is also discussed briefly.

Argenziano and colleagues developed the 7-point checklist in 1998. Two points are assigned to the lesion for each of the major criteria and 1 point for each minor criteria. 

The major criteria include an atypical pigment network, blue-white veil, and atypical vascular pattern; the minor criteria include irregular streaks, irregular pigmentation, irregular dots/globules, and regression structures.11 The lesion shown in Figure 2 scores an 8 out of 10 by this metric, handily surpassing the 3 points required to suggest melanoma.11

The Menzies method was developed by Menzies and colleagues in 1996. To be classified as melanoma, the pigmented lesion must show an absence of pattern symmetry and color uniformity while simultaneously exhibiting at least one of the following: blue-white veil, multiple brown dots, pseudopods, radial streaming, scarlike depigmentation, peripheral block dots/globules, 5 to 6 colors, multiple blue/gray dots, and a broadened network.12 

Again, the lesion shown in Figure 2 meets the criteria concerning for melanoma based on this algorithm.

The ABCD rule is a more technical dermoscopic evaluation algorithm developed in 1994 by Stolz and colleagues. This method yields a numeric value called the total dermoscopic score (TDS) based on Asymmetry, Border pigment pattern, Color variation, and 5 Different structural components. 

The assessment of asymmetry is determined by analyzing the lesion in a plane bisected by 2 axes set at 90°. A score from 0 to 2 is assigned based on the number of axes showing asymmetry in shape, color, or structure. Border pigment pattern is analyzed by dividing the lesion into eighths. A sharp, abrupt change in pigment pattern at the periphery earns the lesion 1 point for each division. The determination of the color variation score is done by adding 1 point for each white, red, light brown, dark brown, blue-gray, or black region identified in the lesion. Last, the lesion is assigned 1 point for each of 5 different structural components observed in the lesion, which include networks, homogenous areas, dots, globules, and streaks. To be significant, homogenous areas must be at least 10% of the lesion, and multiple branched streaks or dots must be visible. The TDS is calculated with the following formula: TDS = 1.3 x Asymmetry + 0.1 x Border + 0.5 x Color + 0.5 x Different. Higher scores are more concerning of melanoma, with scores > 5.45 suggesting melanoma.13 The lesion shown in Figure 2 registers a 7.7 by this metric.

Henning and colleagues developed the CASH algorithm in 2006 with the intention of assisting less experienced dermoscopy users with lesion evaluation.14 This algorithm tallies points for Color, Architectural disorder, Symmetry, and Homogeneity. One point is attributed to a lesion for each light brown, dark brown, black, red, white, and/or blue region present. Architectural disorder is assigned a point value between 0 and 2, with 0 indicating the absence of or minimal architectural disorder, 1 indicating moderate disorder, and 2 indicating marked disorder. Symmetry is assigned a point value between 0 and 2, with 0 points assigned to a lesion that exhibits biaxial symmetry, 1 point assigned to a lesion that exhibits monoaxial symmetry, and 2 points assigned to a lesion that exhibits biaxial asymmetry. Finally, 1 point is attributed to a lesion for evidence of each of the following: atypical network, dots/globules, streaks/pseudopods, blue-white veil, regression structures, blotches > 10% of the overall lesion size, and polymorphous blood vessels. The lesion in Figure 2 scores 16 points out of the maximum total CASH score of 17. Any lesion scoring 8 or more is suggestive of malignant melanoma.14

Finally, the TADA method was developed by Rogers and colleagues in 2016.15 This method uses sequential questions to evaluate lesions. First, “Does the lesion exhibit clear dermoscopic evidence of an angioma, dermatofibroma, or seborrheic keratosis?” If “yes,” then no additional dermoscopic evaluation is necessary, and it is recommended to monitor the lesion. If the answer to the first question is “no,” then ask, “Does the lesion exhibit architectural disorder?” The presence of architectural disorder is based on an overall impression of the lesion, which includes symmetry with regard to structures and colors. Any lesion deemed to exhibit architectural disorder should be biopsied. If the lesion has no architectural disorder, the third question is, “Does the lesion contain any starburst patterns, blue-black or gray coloration, shiny white structures, negative networks, ulcers or erosions, and/or vessels?” If “yes,” then the lesion should be biopsied. Since the lesion in Figure 2 exhibits marked architectural disorder in terms of symmetry and color, analysis of the lesion with the TADA method would yield a recommendation for biopsy.15

 

 

Dermoscopy in Practice

A. Bernard Ackerman, MD, a key figure in the modern era of dermatopathology, wrote an editorial for the Journal of the American Academy of Dermatology in 1985 titled “No one should die of malignant melanoma.” The editorial highlighted that the visual changes associated with melanoma often manifest years prior to malignant invasion and advocated that all physicians should have competence in melanoma detection, specifically mentioning the importance of training primary care physicians (PCPs), dermatologists, and pathologists in this regard.16 This sentiment is equally as true now as it was in 1985.

Naked eye examination paired with an evaluation of patient risk factors for melanoma, including fair skin types, significant sun exposure history, history of sunburn, geographic location, and personal and family history of melanoma, are the foundation of melanoma detection efforts.17 Studies suggest that the triage skills of PCPs could be improved in regard to the evaluation of pigmented lesions. Primary care residents, for instance, did not accurately diagnose 40% of malignant melanoma cases.18,19 Additionally, a meta-analysis demonstrated that PCP accuracy when diagnosing malignant melanoma ranged between 49% and 80%, significantly less than the 85% to 89% exhibited by practicing dermatologists.19 Dermoscopy could be incorporated as an element of the skin examination to enhance lesion discrimination among PCPs, as it has proven use in dermatologic practice.

Dermoscopy is not readily used by PCPs. A survey study of 705 family practitioners in the US performed by Morris and colleagues demonstrated that only 8.3% of participants currently use a dermatoscope to evaluate pigmented lesions.20 The most common barriers to dermoscopy use cited by PCPs in the US include the cost of the dermatoscope, the time required to acquire proficiency, and the lack of financial reimbursement.16 True utilization of dermoscopy among PCPs is higher than this figure suggests due to the number of PCPs who access dermoscopic evaluations via teledermatology. All 21 Veterans Integrated Services Networks of the Veterans Health Administration (VHA) system, for instance, participate in teledermatology and jointly employ more than 1,150 trained telehealth clinical technicians who created a collective 107,000 teledermatology encounters with and without dermoscopy for evaluation by dermatologists in the most recent fiscal year(Martin Weinstock, written communication, October 2017). Nonetheless, it is necessary to determine the contribution that wider utilization of dermoscopy among PCPs would have on melanoma surveillance.

Studies show that dermoscopic algorithms improve the sensitivity while slightly decreasing the specificity of PCPs to detect melanoma compared with that of the naked eye examination. Dolianitis and colleagues demonstrated that a baseline sensitivity of 60.9% for melanoma detection improved to 85.4% with the 7-point checklist, 85.4% with Menzies method, and 77.5% with the ABCD rule, while the baseline specificity of 85.4% moderated to 73.0%, 77.7%, and 80.4%, respectively, among 61 medical practitioners after studying dermoscopy techniques from 2 CDs.21 Westerhoff and colleagues performed a randomized controlled trial with 74 PCPs to determine the effect of a minimal intervention on melanoma diagnostic accuracy. The intervention consisted of providing participants in the experimental group with an atlas of microscopic features common to melanoma to be read at the participants’ leisure, a 1-hour presentation on microscopy, and a 25-questionpractice quiz. Participants randomized to the intervention group improved their diagnostic accuracy from 57.8% to 75.9% with the use of dermoscopy. This group also experiencedimproved accuracy in its clinical diagnosis of melanoma from 54.6% to 62.7%.22

Argenziano and colleagues demonstrated similar results after PCPs attended a 4-hour workshop on dermoscopy. The 73 PCPs in this study evaluated 2,522 lesions randomized to naked eye examination or dermoscopy. The BMR, calculated from the data provided, improved from 12.6:1 to 10.5:1, respectively, when dermoscopy was incorporated into lesion analysis, while the sensitivity increased from 54.1% to 79.2% and the negative predictive value increased from 95.8% to 98.1%. It is important to note that the BMR and negative predictive value improved in tandem, indicating that PCPs were more discriminatory with their referrals for evaluation by dermatology while capturing a greater percentage of melanomas.23

These studies are not without limitations that preclude broad generalizations. For example, Dolianitis and colleagues and Westerhoff and colleagues provided participants with dermoscopic images of the lesions to be evaluated instead of requiring personal use of a dermatoscope, whereas the study by Argenziano and colleagues incorporated only 6 histopathologically proven malignant melanomas into each of the naked eye examination and the experimental dermoscopy groups.21-23 Yet these studies suggest that broader use of dermoscopy among PCPs could improve the accuracy of melanoma detection given clinically relevant training.

Several additional studies identify positive correlations associated with dermoscopy use among PCPs. A recent survey of 425 French general practitioners found that 8% of the study participants acknowledged owning a dermatoscope. Dermatoscope owners spent a statistically significant longer time analyzing each pigmented skin lesions, exhibited greater confidence in their analysis of pigmented lesions, and issued fewer overall referrals to dermatologists.24

Similarly, Rosendahl and colleagues evaluated the number needed to treat (NNT) (equivalent to the BMR) among 193 Australian PCPs and found that the NNT was inversely correlated to the frequency with which the physicians used dermoscopy. However, it was difficult to determine the definitive cause of the reduced NNT in this study because a similar effect was observed when NNT was evaluated based on general practitioner subspecialization.25 Again, despite limitations, these studies suggest that increased dermoscopy use among PCPs could reduce the morbidity of lifelong scarring as well as the short-term anxiety associated with a possible melanoma diagnosis.

 

 

Limitations

Even in the hands of a trained dermatologist, dermoscopy has limitations. Featureless melanoma is a term applied to melanoma lesions bereft of classical findings on both naked eye examination and dermoscopy. Menzies, a dermatologic pioneer in dermoscopy, acknowledged this limitation in 1996 while showing that 8% of melanomas evaded dermoscopic detection. He proceeded to discuss the importance of clinical history in melanoma detection because all of the featureless melanomas exhibited recent changes in size, shape, and/or color.26 More recently, sequential dermoscopy (mole mapping) imaging has been implemented to successfully identify these lesions.27 Thus, dermoscopy cannot replace dermatologists trained in the art of visual assessment with honed clinical diagnostic acumen. Rather, dermoscopy is a tool to enhance the assessment of clinically suspicious lesions and aid diagnostic discrimination of uncertain pigmented lesions.

Conclusion

Primary care physicians are on the frontline of medicine and often the first to have the opportunity to detect the presence of melanoma. Notably, 52.2% of the 884.7 million medical office visits performed annually in the US are with PCPs.28 Despite the benefits, dermoscopy is not uniformly used by dermatologists in the US. Of dermatologists practicing for more than 20 years, 76.2% use dermoscopy compared with 97.8% of dermatologists in practice for less than 5 years. This illustrates an increased use in tandem with dermatology residencies integrating dermoscopy training as a component of the curriculum, showing the importance of incorporating dermoscopy into medical school and residency training for PCPs..29-31 Guidelines regarding dermoscopy training and dermoscopic evaluation algorithms should be established, routinely taught in medical education, and actively incorporated into training curriculum for PCPs in order to improve patient care and realize the potential health care savings associated with the early diagnosis and treatment of melanoma. Dermoscopic-teledermatology consultations present a viable opportunity within the VHA to expedite access to care for veterans and simultaneously offer evaluative feedback on lesions to referring PCPs, as skilled, dermoscopy-trained dermatologists render the diagnoses. Given the devastating mortality rate of melanoma, continued multidisciplinary education on identifying melanoma is of the utmost importance for patient care. Widespread implementation of dermoscopy and dermoscopic-teledermatology consultations could save lives and slow the ever-increasing economic burden associated with melanoma treatment, costing $1.467 billion in 2016.32

References

1. Guy GP Jr, Thomas CC, Thompson T, Watson M, Massetti GM, Richardson LC. Vital signs: melanoma incidence and mortality trends and projections-United States, 1982-2030. MMWR Morb Mortal Wkly Rep. 2015;64(21):591-596.

2. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J Clin. 2018;68(1):7-30.

3. American Cancer Society. Cancer facts & figures 2017. Atlanta: American Cancer Society; 2017. https://www.cancer.org/content/dam/cancer-org/research/cancer-facts-and-statistics/annual-cancer-facts-and-figures/2017/cancer-facts-and-figures-2017.pdf. Accessed April 19, 2018.

4. Lea CS, Efird JT, Toland AE, Lewis DR, Phillips CJ. Melanoma incidence rates in active duty military personnel compared with a population-based registry in the United States, 2000-2007. Mil Med. 2014;179(3):247-253.

5. Thomas L, Puig S. Dermoscopy, digital dermoscopy and other diagnostic tools in the early detection of melanoma and follow-up of high-risk skin cancer patients. Acta Derm Venereol. 2017;97(218):14-21.

6. Marghoob AA, Usatine RP, Jaimes N. Dermoscopy for the family physician. Am Fam Physician. 2013;88(7):441-450.

7. Bafounta ML, Beauchet A, Aegerter P, Saiag P. Is dermoscopy (epiluminescence microscopy) useful for the diagnosis of melanoma? Results of a meta-analysis using techniques adapted to the evaluation of diagnostic tests. Arch Dermatol. 2001;137(10):1343-1350.

8. Terushkin V, Warycha M, Levy M, Kopf AW, Cohen DE, Polsky D. Analysis of the benign to malignant ratio of lesions biopsied by a general dermatologist before and after the adoption of dermoscopy. Arch Dermatol. 2010;146(3):343-344.

9. Wolner ZJ, Yélamos O, Liopyris K, Rogers T, Marchetti MA, Marghoob AA. Enhancing skin cancer diagnosis with dermoscopy. Dermatol Clin. 2017;35(4):417-437.

10. Carli P, Quercioli E, Sestini S, et al. Pattern analysis, not simplified algorithms, is the most reliable method for teaching dermoscopy for melanoma diagnosis to residents in dermatology. Br J Dermatol. 2003;148(5):981-984.

11. Argenziano G, Fabbrocini G, Carli P, De Giorgi V, Sammarco E, Delfino M. Epiluminescence microscopy for the diagnosis of doubtful melanocytic skin lesions. Comparison of the ABCD rule of dermatoscopy and a new 7-point checklist based on pattern analysis. Arch Dermatol. 1998;134(12):1563-1570.

12. Menzies SW, Ingvar C, Crotty KA, McCarthy WH. Frequency and morphologic characteristics of invasive melanomas lacking specific surface microscopic features. Arch Dermatol. 1996;132(10):1178-1182.

13. Nachbar F, Stolz W, Merkle T, et al. The ABCD rule of dermatoscopy. High prospective value in the diagnosis of doubtful melanocytic skin lesions. J Am Acad Dermatol. 1994;30(4):551-559.

14. Henning JS, Dusza SW, Wang SQ, et al. The CASH (color, architecture, symmetry, and homogeneity) algorithm for dermoscopy. J Am Acad Dermatol. 2007;56(1):45-52.

15. Rogers T, Marino M, Dusza SW, Bajaj S, Marchetti MA, Marghoob A. Triage amalgamated dermoscopic algorithm (TADA) for skin cancer screening. Dermatol Pract Concept. 2017;7(2):39-46.

16. Ackerman AB. No one should die of malignant melanoma. J Am Acad Dermatol. 1985;12(1):115-116.

17. Gandini S, Sera F, Cattaruzza MS, et al. Meta-analysis of risk factors for cutaneous melanoma: II: sun exposure. Eur J Cancer. 2005;41(1):45-60.

18. Gerbert B, Maurer T, Berger T, et al. Primary care physicians as gatekeepers in managed care. Primary care physicians’ and dermatologists’ skills at secondary prevention of skin cancer. Arch Dermatol. 1996;132(9):1030-1038.

19. Corbo MD, Wismer J. Agreement between dermatologists and primary care practitioners in the diagnosis of malignant melanoma: review of the literature. J Cutan Med Surg. 2012;16(5):306-310.

20. Morris JB, Alfonso SV, Hernandez N, Fernández MI. Examining the factors associated with past and present dermoscopy use among family physicians. Dermatol Pract Concept. 2017;7(4):63-70.

21. Dolianitis C, Kelly J, Wolfe R, Simpson P. Comparative performance of 4 dermoscopic algorithms by nonexperts for the diagnosis of melanocytic lesions. Arch Dermatol. 2005;141(8):1008-1014.

22. Westerhoff K, Mccarthy WH, Menzies SW. Increase in the sensitivity for melanoma diagnosis by primary care physicians using skin surface microscopy. Br J Dermatol. 2000;143(5):1016-1020.

23. Argenziano G, Puig S, Zalaudek I, et al. Dermoscopy improves accuracy of primary care physicians to triage lesions suggestive of skin cancer. J Clin Oncol. 2006;24(12):1877-1882.

24. Chappuis P, Duru G, Marchal O, Girier P, Dalle S, Thomas L. Dermoscopy, a useful tool for general practitioners in melanoma screening: a nationwide survey. Br J Dermatol. 2016;175(4):744-750.

25. Rosendahl C, Williams G, Eley D, et al. The impact of subspecialization and dermatoscopy use on accuracy of melanoma diagnosis among primary care doctors in Australia. J Am Acad Dermatol. 2012;67(5):846-852.

26. Menzies SW, Ingvar C, Crotty KA, McCarthy WH. Frequency and morphologic characteristics of invasive melanomas lacking specific surface microscopic features. Arch Dermatol. 1996;132(10):1178-1182.

27. Kittler H, Guitera P, Riedl E, et al. Identification of clinically featureless incipient melanoma using sequential dermoscopy imaging. Arch Dermatol. 2006;142(9):1113-1119.

28. Centers for Disease Control and Prevention. Ambulatory care use and physician office visits. https://www.cdc.gov/nchs/fastats/physician-visits.htm. Updated May 3, 2017. Accessed April 10, 2018.

29. Murzaku EC, Hayan S, Rao BK. Methods and rates of dermoscopy usage: a cross-sectional survey of US dermatologists stratified by years in practice. J Am Acad Dermatol. 2014;71(2):393-395.

30. Nehal KS, Oliveria SA, Marghoob AA, et al. Use of and beliefs about dermoscopy in the management of patients with pigmented lesions: a survey of dermatology residency programmes in the United States. Melanoma Res. 2002;12(6):601-605.

31. Wu TP, Newlove T, Smith L, Vuong CH, Stein JA, Polsky D. The importance of dedicated dermoscopy training during residency: a survey of US dermatology chief residents. J Am Acad Dermatol. 2013;68(6):1000-1005.

32. Lim HW, Collins SAB, Resneck JS Jr, et al. The burden of skin disease in the United States. J Am Acad Dermatol. 2017;76(5):958-972

Article PDF
Author and Disclosure Information

Mr. Holmes is a Medical Student at the University of California, Riverside in Riverside, California. Ms. Limone is a Medical Student, Dr. Vassantachart is a PGY-2 Dermatology Resident, and Dr. Zumwalt is a PGY-4 Dermatology Resident, all at Loma Linda University in Loma Linda, California. Dr. Jacob is the Dermatology Section Chief and Professor of Dermatology, and Dr. Hirokane is the Dermatology Clinical Director
and Assistant Professor of Dermatology, both at Loma Linda University and the Loma Linda VA Medical Center.
Correspondence: Dr. Jacob (sharon.jacob@va.gov)

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

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

Issue
Federal Practitioner - 35(4)s
Publications
Topics
Page Number
S39-S45
Sections
Author and Disclosure Information

Mr. Holmes is a Medical Student at the University of California, Riverside in Riverside, California. Ms. Limone is a Medical Student, Dr. Vassantachart is a PGY-2 Dermatology Resident, and Dr. Zumwalt is a PGY-4 Dermatology Resident, all at Loma Linda University in Loma Linda, California. Dr. Jacob is the Dermatology Section Chief and Professor of Dermatology, and Dr. Hirokane is the Dermatology Clinical Director
and Assistant Professor of Dermatology, both at Loma Linda University and the Loma Linda VA Medical Center.
Correspondence: Dr. Jacob (sharon.jacob@va.gov)

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

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

Author and Disclosure Information

Mr. Holmes is a Medical Student at the University of California, Riverside in Riverside, California. Ms. Limone is a Medical Student, Dr. Vassantachart is a PGY-2 Dermatology Resident, and Dr. Zumwalt is a PGY-4 Dermatology Resident, all at Loma Linda University in Loma Linda, California. Dr. Jacob is the Dermatology Section Chief and Professor of Dermatology, and Dr. Hirokane is the Dermatology Clinical Director
and Assistant Professor of Dermatology, both at Loma Linda University and the Loma Linda VA Medical Center.
Correspondence: Dr. Jacob (sharon.jacob@va.gov)

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

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

Article PDF
Article PDF
Use of dermoscopy and detection algorithms by primary care physicians can enhance assessment of clinically suspicious lesions compared with that of naked eye examinations.
Use of dermoscopy and detection algorithms by primary care physicians can enhance assessment of clinically suspicious lesions compared with that of naked eye examinations.

From 1982 to 2011, the melanoma incidence rate doubled in the US.1 In 2018, an estimated 87,290 cases of melanoma in situ and 91,270 cases of invasive melanoma will be diagnosed in the US, and 9,320 deaths will be attributable to melanoma.2 Early detection of melanoma is critically important to reduce melanoma-related mortality, with 5-year survival rates as high as 97% at stage 1A vs a 20% 5-year survival when there is distant metastasis.2,3 Melanoma is particularly relevant for medical providers working with veterans because melanoma disproportionately affects service members with an incidence rate ratio of 1.62 (95% confidence interval [CI], 1.40-1.86) compared with that of the general population.4

Biopsy is the definitive diagnostic tool for melanoma. Histologic analysis differentiates melanoma from seborrheic keratoses, pigmented nevi, dermatofibromas, and other pigmented lesions that can resemble melanoma on clinical examination. However, biopsy must be used judiciously as unnecessary biopsies contribute to health care costs and leave scars, which can have psychosocial implications. With benign nevi outnumbering melanoma about 2 million to 1, biopsy is indicated once a threshold of suspicion is obtained.5

Dermoscopic Tool

Dermoscopy is a microscopy-based tool to improve noninvasive diagnostic discrimination of skin lesions based on color and structure analysis. Coloration provides an indication of the composition of elements present in the skin with keratin appearing yellow, blood appearing red, and collagen appearing white. Coloration also suggests pigment depth as melanin appears black when located in the stratum corneum, brown when located deeper in the epidermis, and blue when located in the dermis.6 Finally, characteristic histopathologic alterations in the dermoepidermal junction, rete ridges, pigment-containing cells, and/or melanocyte granules that occur in melanoma are recognizable with dermoscopy.6

In 2001, Bafounta and colleagues performed the first meta-analysis on the efficacy of dermoscopy compared with that of clinical evaluation, finding that dermoscopy performed specifically by dermatology-trained clinicians improved the accuracy of identifying melanoma from an odds ratio of 16 (95% CI, 9-31) with naked eye examination to 76 (95% CI, 25-223) with dermoscopy.7

More recently, Terushkin and colleagues demonstrated that diagnosis specificity improves when a general dermatologist is trained in dermoscopic pattern recognition. Naked eye examination produced a benign to malignant ratio (BMR) of 18.4:1, indicating that about 18 of 19 biopsies considered suspicious for melanoma ultimately yielded benign melanocytic lesions. Although the BMR for the general dermatologist experienced an increase after dermoscopy training, the ratio eventually decreased to 7.9:1.8

Dermoscopic Analysis

Some of the common patterns recognized in melanoma are demonstrated in Figures 1 and 2. Figure 1 is a close-up of a patient’s upper back showing a solitary asymmetric melanocytic lesion containing multiple red, brown, black, and blue hues. 

The lesion is highly suspicious for melanoma. Key patterns identified under dermoscopy in Figure 2 increase the level of suspicion. The pink circle in the upper left of the lesion demonstrates a scarlike regression of pigment structure. 
The orange triangle signifies a region with marked variability in color called an atypical pigment network, and the centrally located yellow circle and gray square identify interspersed atypical dots and globules of color. The red rectangle on the right highlights irregular streaking, linear radial projections suggestive of superficial spreading melanoma. The green line identifies hypopigmentation with surrounding curvilinear globular structures collectively known as a negative network.  Finally, the bottom blue triangle overlies an area with a hazy blue tinge called a blue-white veil, indicating the presence of melanocytes deep in the dermis (Table 1).6,9

Pattern analysis, the dermoscopic interpretation method preferred by pigmented lesion specialists, requires simultaneously assessing numerous lesion patterns that vary depending on body site.10 Alternative dermoscopic algorithms that focus on the most common features of melanoma have been developed to aid practitioners with the interpretation of dermoscopy findings: the 7-point checklist, the Menzies method, the ABCD rule, and the CASH algorithm (Tables 2, 3, 4, and 5). 

To apply these algorithms to evaluate the lesion in Figures 1 and 2 ( eAppendix ).11-14 The triage amalgamated dermoscopic algorithm (TADA) method, a newer algorithm designed for novice dermoscopy users, is also discussed briefly.

Argenziano and colleagues developed the 7-point checklist in 1998. Two points are assigned to the lesion for each of the major criteria and 1 point for each minor criteria. 

The major criteria include an atypical pigment network, blue-white veil, and atypical vascular pattern; the minor criteria include irregular streaks, irregular pigmentation, irregular dots/globules, and regression structures.11 The lesion shown in Figure 2 scores an 8 out of 10 by this metric, handily surpassing the 3 points required to suggest melanoma.11

The Menzies method was developed by Menzies and colleagues in 1996. To be classified as melanoma, the pigmented lesion must show an absence of pattern symmetry and color uniformity while simultaneously exhibiting at least one of the following: blue-white veil, multiple brown dots, pseudopods, radial streaming, scarlike depigmentation, peripheral block dots/globules, 5 to 6 colors, multiple blue/gray dots, and a broadened network.12 

Again, the lesion shown in Figure 2 meets the criteria concerning for melanoma based on this algorithm.

The ABCD rule is a more technical dermoscopic evaluation algorithm developed in 1994 by Stolz and colleagues. This method yields a numeric value called the total dermoscopic score (TDS) based on Asymmetry, Border pigment pattern, Color variation, and 5 Different structural components. 

The assessment of asymmetry is determined by analyzing the lesion in a plane bisected by 2 axes set at 90°. A score from 0 to 2 is assigned based on the number of axes showing asymmetry in shape, color, or structure. Border pigment pattern is analyzed by dividing the lesion into eighths. A sharp, abrupt change in pigment pattern at the periphery earns the lesion 1 point for each division. The determination of the color variation score is done by adding 1 point for each white, red, light brown, dark brown, blue-gray, or black region identified in the lesion. Last, the lesion is assigned 1 point for each of 5 different structural components observed in the lesion, which include networks, homogenous areas, dots, globules, and streaks. To be significant, homogenous areas must be at least 10% of the lesion, and multiple branched streaks or dots must be visible. The TDS is calculated with the following formula: TDS = 1.3 x Asymmetry + 0.1 x Border + 0.5 x Color + 0.5 x Different. Higher scores are more concerning of melanoma, with scores > 5.45 suggesting melanoma.13 The lesion shown in Figure 2 registers a 7.7 by this metric.

Henning and colleagues developed the CASH algorithm in 2006 with the intention of assisting less experienced dermoscopy users with lesion evaluation.14 This algorithm tallies points for Color, Architectural disorder, Symmetry, and Homogeneity. One point is attributed to a lesion for each light brown, dark brown, black, red, white, and/or blue region present. Architectural disorder is assigned a point value between 0 and 2, with 0 indicating the absence of or minimal architectural disorder, 1 indicating moderate disorder, and 2 indicating marked disorder. Symmetry is assigned a point value between 0 and 2, with 0 points assigned to a lesion that exhibits biaxial symmetry, 1 point assigned to a lesion that exhibits monoaxial symmetry, and 2 points assigned to a lesion that exhibits biaxial asymmetry. Finally, 1 point is attributed to a lesion for evidence of each of the following: atypical network, dots/globules, streaks/pseudopods, blue-white veil, regression structures, blotches > 10% of the overall lesion size, and polymorphous blood vessels. The lesion in Figure 2 scores 16 points out of the maximum total CASH score of 17. Any lesion scoring 8 or more is suggestive of malignant melanoma.14

Finally, the TADA method was developed by Rogers and colleagues in 2016.15 This method uses sequential questions to evaluate lesions. First, “Does the lesion exhibit clear dermoscopic evidence of an angioma, dermatofibroma, or seborrheic keratosis?” If “yes,” then no additional dermoscopic evaluation is necessary, and it is recommended to monitor the lesion. If the answer to the first question is “no,” then ask, “Does the lesion exhibit architectural disorder?” The presence of architectural disorder is based on an overall impression of the lesion, which includes symmetry with regard to structures and colors. Any lesion deemed to exhibit architectural disorder should be biopsied. If the lesion has no architectural disorder, the third question is, “Does the lesion contain any starburst patterns, blue-black or gray coloration, shiny white structures, negative networks, ulcers or erosions, and/or vessels?” If “yes,” then the lesion should be biopsied. Since the lesion in Figure 2 exhibits marked architectural disorder in terms of symmetry and color, analysis of the lesion with the TADA method would yield a recommendation for biopsy.15

 

 

Dermoscopy in Practice

A. Bernard Ackerman, MD, a key figure in the modern era of dermatopathology, wrote an editorial for the Journal of the American Academy of Dermatology in 1985 titled “No one should die of malignant melanoma.” The editorial highlighted that the visual changes associated with melanoma often manifest years prior to malignant invasion and advocated that all physicians should have competence in melanoma detection, specifically mentioning the importance of training primary care physicians (PCPs), dermatologists, and pathologists in this regard.16 This sentiment is equally as true now as it was in 1985.

Naked eye examination paired with an evaluation of patient risk factors for melanoma, including fair skin types, significant sun exposure history, history of sunburn, geographic location, and personal and family history of melanoma, are the foundation of melanoma detection efforts.17 Studies suggest that the triage skills of PCPs could be improved in regard to the evaluation of pigmented lesions. Primary care residents, for instance, did not accurately diagnose 40% of malignant melanoma cases.18,19 Additionally, a meta-analysis demonstrated that PCP accuracy when diagnosing malignant melanoma ranged between 49% and 80%, significantly less than the 85% to 89% exhibited by practicing dermatologists.19 Dermoscopy could be incorporated as an element of the skin examination to enhance lesion discrimination among PCPs, as it has proven use in dermatologic practice.

Dermoscopy is not readily used by PCPs. A survey study of 705 family practitioners in the US performed by Morris and colleagues demonstrated that only 8.3% of participants currently use a dermatoscope to evaluate pigmented lesions.20 The most common barriers to dermoscopy use cited by PCPs in the US include the cost of the dermatoscope, the time required to acquire proficiency, and the lack of financial reimbursement.16 True utilization of dermoscopy among PCPs is higher than this figure suggests due to the number of PCPs who access dermoscopic evaluations via teledermatology. All 21 Veterans Integrated Services Networks of the Veterans Health Administration (VHA) system, for instance, participate in teledermatology and jointly employ more than 1,150 trained telehealth clinical technicians who created a collective 107,000 teledermatology encounters with and without dermoscopy for evaluation by dermatologists in the most recent fiscal year(Martin Weinstock, written communication, October 2017). Nonetheless, it is necessary to determine the contribution that wider utilization of dermoscopy among PCPs would have on melanoma surveillance.

Studies show that dermoscopic algorithms improve the sensitivity while slightly decreasing the specificity of PCPs to detect melanoma compared with that of the naked eye examination. Dolianitis and colleagues demonstrated that a baseline sensitivity of 60.9% for melanoma detection improved to 85.4% with the 7-point checklist, 85.4% with Menzies method, and 77.5% with the ABCD rule, while the baseline specificity of 85.4% moderated to 73.0%, 77.7%, and 80.4%, respectively, among 61 medical practitioners after studying dermoscopy techniques from 2 CDs.21 Westerhoff and colleagues performed a randomized controlled trial with 74 PCPs to determine the effect of a minimal intervention on melanoma diagnostic accuracy. The intervention consisted of providing participants in the experimental group with an atlas of microscopic features common to melanoma to be read at the participants’ leisure, a 1-hour presentation on microscopy, and a 25-questionpractice quiz. Participants randomized to the intervention group improved their diagnostic accuracy from 57.8% to 75.9% with the use of dermoscopy. This group also experiencedimproved accuracy in its clinical diagnosis of melanoma from 54.6% to 62.7%.22

Argenziano and colleagues demonstrated similar results after PCPs attended a 4-hour workshop on dermoscopy. The 73 PCPs in this study evaluated 2,522 lesions randomized to naked eye examination or dermoscopy. The BMR, calculated from the data provided, improved from 12.6:1 to 10.5:1, respectively, when dermoscopy was incorporated into lesion analysis, while the sensitivity increased from 54.1% to 79.2% and the negative predictive value increased from 95.8% to 98.1%. It is important to note that the BMR and negative predictive value improved in tandem, indicating that PCPs were more discriminatory with their referrals for evaluation by dermatology while capturing a greater percentage of melanomas.23

These studies are not without limitations that preclude broad generalizations. For example, Dolianitis and colleagues and Westerhoff and colleagues provided participants with dermoscopic images of the lesions to be evaluated instead of requiring personal use of a dermatoscope, whereas the study by Argenziano and colleagues incorporated only 6 histopathologically proven malignant melanomas into each of the naked eye examination and the experimental dermoscopy groups.21-23 Yet these studies suggest that broader use of dermoscopy among PCPs could improve the accuracy of melanoma detection given clinically relevant training.

Several additional studies identify positive correlations associated with dermoscopy use among PCPs. A recent survey of 425 French general practitioners found that 8% of the study participants acknowledged owning a dermatoscope. Dermatoscope owners spent a statistically significant longer time analyzing each pigmented skin lesions, exhibited greater confidence in their analysis of pigmented lesions, and issued fewer overall referrals to dermatologists.24

Similarly, Rosendahl and colleagues evaluated the number needed to treat (NNT) (equivalent to the BMR) among 193 Australian PCPs and found that the NNT was inversely correlated to the frequency with which the physicians used dermoscopy. However, it was difficult to determine the definitive cause of the reduced NNT in this study because a similar effect was observed when NNT was evaluated based on general practitioner subspecialization.25 Again, despite limitations, these studies suggest that increased dermoscopy use among PCPs could reduce the morbidity of lifelong scarring as well as the short-term anxiety associated with a possible melanoma diagnosis.

 

 

Limitations

Even in the hands of a trained dermatologist, dermoscopy has limitations. Featureless melanoma is a term applied to melanoma lesions bereft of classical findings on both naked eye examination and dermoscopy. Menzies, a dermatologic pioneer in dermoscopy, acknowledged this limitation in 1996 while showing that 8% of melanomas evaded dermoscopic detection. He proceeded to discuss the importance of clinical history in melanoma detection because all of the featureless melanomas exhibited recent changes in size, shape, and/or color.26 More recently, sequential dermoscopy (mole mapping) imaging has been implemented to successfully identify these lesions.27 Thus, dermoscopy cannot replace dermatologists trained in the art of visual assessment with honed clinical diagnostic acumen. Rather, dermoscopy is a tool to enhance the assessment of clinically suspicious lesions and aid diagnostic discrimination of uncertain pigmented lesions.

Conclusion

Primary care physicians are on the frontline of medicine and often the first to have the opportunity to detect the presence of melanoma. Notably, 52.2% of the 884.7 million medical office visits performed annually in the US are with PCPs.28 Despite the benefits, dermoscopy is not uniformly used by dermatologists in the US. Of dermatologists practicing for more than 20 years, 76.2% use dermoscopy compared with 97.8% of dermatologists in practice for less than 5 years. This illustrates an increased use in tandem with dermatology residencies integrating dermoscopy training as a component of the curriculum, showing the importance of incorporating dermoscopy into medical school and residency training for PCPs..29-31 Guidelines regarding dermoscopy training and dermoscopic evaluation algorithms should be established, routinely taught in medical education, and actively incorporated into training curriculum for PCPs in order to improve patient care and realize the potential health care savings associated with the early diagnosis and treatment of melanoma. Dermoscopic-teledermatology consultations present a viable opportunity within the VHA to expedite access to care for veterans and simultaneously offer evaluative feedback on lesions to referring PCPs, as skilled, dermoscopy-trained dermatologists render the diagnoses. Given the devastating mortality rate of melanoma, continued multidisciplinary education on identifying melanoma is of the utmost importance for patient care. Widespread implementation of dermoscopy and dermoscopic-teledermatology consultations could save lives and slow the ever-increasing economic burden associated with melanoma treatment, costing $1.467 billion in 2016.32

From 1982 to 2011, the melanoma incidence rate doubled in the US.1 In 2018, an estimated 87,290 cases of melanoma in situ and 91,270 cases of invasive melanoma will be diagnosed in the US, and 9,320 deaths will be attributable to melanoma.2 Early detection of melanoma is critically important to reduce melanoma-related mortality, with 5-year survival rates as high as 97% at stage 1A vs a 20% 5-year survival when there is distant metastasis.2,3 Melanoma is particularly relevant for medical providers working with veterans because melanoma disproportionately affects service members with an incidence rate ratio of 1.62 (95% confidence interval [CI], 1.40-1.86) compared with that of the general population.4

Biopsy is the definitive diagnostic tool for melanoma. Histologic analysis differentiates melanoma from seborrheic keratoses, pigmented nevi, dermatofibromas, and other pigmented lesions that can resemble melanoma on clinical examination. However, biopsy must be used judiciously as unnecessary biopsies contribute to health care costs and leave scars, which can have psychosocial implications. With benign nevi outnumbering melanoma about 2 million to 1, biopsy is indicated once a threshold of suspicion is obtained.5

Dermoscopic Tool

Dermoscopy is a microscopy-based tool to improve noninvasive diagnostic discrimination of skin lesions based on color and structure analysis. Coloration provides an indication of the composition of elements present in the skin with keratin appearing yellow, blood appearing red, and collagen appearing white. Coloration also suggests pigment depth as melanin appears black when located in the stratum corneum, brown when located deeper in the epidermis, and blue when located in the dermis.6 Finally, characteristic histopathologic alterations in the dermoepidermal junction, rete ridges, pigment-containing cells, and/or melanocyte granules that occur in melanoma are recognizable with dermoscopy.6

In 2001, Bafounta and colleagues performed the first meta-analysis on the efficacy of dermoscopy compared with that of clinical evaluation, finding that dermoscopy performed specifically by dermatology-trained clinicians improved the accuracy of identifying melanoma from an odds ratio of 16 (95% CI, 9-31) with naked eye examination to 76 (95% CI, 25-223) with dermoscopy.7

More recently, Terushkin and colleagues demonstrated that diagnosis specificity improves when a general dermatologist is trained in dermoscopic pattern recognition. Naked eye examination produced a benign to malignant ratio (BMR) of 18.4:1, indicating that about 18 of 19 biopsies considered suspicious for melanoma ultimately yielded benign melanocytic lesions. Although the BMR for the general dermatologist experienced an increase after dermoscopy training, the ratio eventually decreased to 7.9:1.8

Dermoscopic Analysis

Some of the common patterns recognized in melanoma are demonstrated in Figures 1 and 2. Figure 1 is a close-up of a patient’s upper back showing a solitary asymmetric melanocytic lesion containing multiple red, brown, black, and blue hues. 

The lesion is highly suspicious for melanoma. Key patterns identified under dermoscopy in Figure 2 increase the level of suspicion. The pink circle in the upper left of the lesion demonstrates a scarlike regression of pigment structure. 
The orange triangle signifies a region with marked variability in color called an atypical pigment network, and the centrally located yellow circle and gray square identify interspersed atypical dots and globules of color. The red rectangle on the right highlights irregular streaking, linear radial projections suggestive of superficial spreading melanoma. The green line identifies hypopigmentation with surrounding curvilinear globular structures collectively known as a negative network.  Finally, the bottom blue triangle overlies an area with a hazy blue tinge called a blue-white veil, indicating the presence of melanocytes deep in the dermis (Table 1).6,9

Pattern analysis, the dermoscopic interpretation method preferred by pigmented lesion specialists, requires simultaneously assessing numerous lesion patterns that vary depending on body site.10 Alternative dermoscopic algorithms that focus on the most common features of melanoma have been developed to aid practitioners with the interpretation of dermoscopy findings: the 7-point checklist, the Menzies method, the ABCD rule, and the CASH algorithm (Tables 2, 3, 4, and 5). 

To apply these algorithms to evaluate the lesion in Figures 1 and 2 ( eAppendix ).11-14 The triage amalgamated dermoscopic algorithm (TADA) method, a newer algorithm designed for novice dermoscopy users, is also discussed briefly.

Argenziano and colleagues developed the 7-point checklist in 1998. Two points are assigned to the lesion for each of the major criteria and 1 point for each minor criteria. 

The major criteria include an atypical pigment network, blue-white veil, and atypical vascular pattern; the minor criteria include irregular streaks, irregular pigmentation, irregular dots/globules, and regression structures.11 The lesion shown in Figure 2 scores an 8 out of 10 by this metric, handily surpassing the 3 points required to suggest melanoma.11

The Menzies method was developed by Menzies and colleagues in 1996. To be classified as melanoma, the pigmented lesion must show an absence of pattern symmetry and color uniformity while simultaneously exhibiting at least one of the following: blue-white veil, multiple brown dots, pseudopods, radial streaming, scarlike depigmentation, peripheral block dots/globules, 5 to 6 colors, multiple blue/gray dots, and a broadened network.12 

Again, the lesion shown in Figure 2 meets the criteria concerning for melanoma based on this algorithm.

The ABCD rule is a more technical dermoscopic evaluation algorithm developed in 1994 by Stolz and colleagues. This method yields a numeric value called the total dermoscopic score (TDS) based on Asymmetry, Border pigment pattern, Color variation, and 5 Different structural components. 

The assessment of asymmetry is determined by analyzing the lesion in a plane bisected by 2 axes set at 90°. A score from 0 to 2 is assigned based on the number of axes showing asymmetry in shape, color, or structure. Border pigment pattern is analyzed by dividing the lesion into eighths. A sharp, abrupt change in pigment pattern at the periphery earns the lesion 1 point for each division. The determination of the color variation score is done by adding 1 point for each white, red, light brown, dark brown, blue-gray, or black region identified in the lesion. Last, the lesion is assigned 1 point for each of 5 different structural components observed in the lesion, which include networks, homogenous areas, dots, globules, and streaks. To be significant, homogenous areas must be at least 10% of the lesion, and multiple branched streaks or dots must be visible. The TDS is calculated with the following formula: TDS = 1.3 x Asymmetry + 0.1 x Border + 0.5 x Color + 0.5 x Different. Higher scores are more concerning of melanoma, with scores > 5.45 suggesting melanoma.13 The lesion shown in Figure 2 registers a 7.7 by this metric.

Henning and colleagues developed the CASH algorithm in 2006 with the intention of assisting less experienced dermoscopy users with lesion evaluation.14 This algorithm tallies points for Color, Architectural disorder, Symmetry, and Homogeneity. One point is attributed to a lesion for each light brown, dark brown, black, red, white, and/or blue region present. Architectural disorder is assigned a point value between 0 and 2, with 0 indicating the absence of or minimal architectural disorder, 1 indicating moderate disorder, and 2 indicating marked disorder. Symmetry is assigned a point value between 0 and 2, with 0 points assigned to a lesion that exhibits biaxial symmetry, 1 point assigned to a lesion that exhibits monoaxial symmetry, and 2 points assigned to a lesion that exhibits biaxial asymmetry. Finally, 1 point is attributed to a lesion for evidence of each of the following: atypical network, dots/globules, streaks/pseudopods, blue-white veil, regression structures, blotches > 10% of the overall lesion size, and polymorphous blood vessels. The lesion in Figure 2 scores 16 points out of the maximum total CASH score of 17. Any lesion scoring 8 or more is suggestive of malignant melanoma.14

Finally, the TADA method was developed by Rogers and colleagues in 2016.15 This method uses sequential questions to evaluate lesions. First, “Does the lesion exhibit clear dermoscopic evidence of an angioma, dermatofibroma, or seborrheic keratosis?” If “yes,” then no additional dermoscopic evaluation is necessary, and it is recommended to monitor the lesion. If the answer to the first question is “no,” then ask, “Does the lesion exhibit architectural disorder?” The presence of architectural disorder is based on an overall impression of the lesion, which includes symmetry with regard to structures and colors. Any lesion deemed to exhibit architectural disorder should be biopsied. If the lesion has no architectural disorder, the third question is, “Does the lesion contain any starburst patterns, blue-black or gray coloration, shiny white structures, negative networks, ulcers or erosions, and/or vessels?” If “yes,” then the lesion should be biopsied. Since the lesion in Figure 2 exhibits marked architectural disorder in terms of symmetry and color, analysis of the lesion with the TADA method would yield a recommendation for biopsy.15

 

 

Dermoscopy in Practice

A. Bernard Ackerman, MD, a key figure in the modern era of dermatopathology, wrote an editorial for the Journal of the American Academy of Dermatology in 1985 titled “No one should die of malignant melanoma.” The editorial highlighted that the visual changes associated with melanoma often manifest years prior to malignant invasion and advocated that all physicians should have competence in melanoma detection, specifically mentioning the importance of training primary care physicians (PCPs), dermatologists, and pathologists in this regard.16 This sentiment is equally as true now as it was in 1985.

Naked eye examination paired with an evaluation of patient risk factors for melanoma, including fair skin types, significant sun exposure history, history of sunburn, geographic location, and personal and family history of melanoma, are the foundation of melanoma detection efforts.17 Studies suggest that the triage skills of PCPs could be improved in regard to the evaluation of pigmented lesions. Primary care residents, for instance, did not accurately diagnose 40% of malignant melanoma cases.18,19 Additionally, a meta-analysis demonstrated that PCP accuracy when diagnosing malignant melanoma ranged between 49% and 80%, significantly less than the 85% to 89% exhibited by practicing dermatologists.19 Dermoscopy could be incorporated as an element of the skin examination to enhance lesion discrimination among PCPs, as it has proven use in dermatologic practice.

Dermoscopy is not readily used by PCPs. A survey study of 705 family practitioners in the US performed by Morris and colleagues demonstrated that only 8.3% of participants currently use a dermatoscope to evaluate pigmented lesions.20 The most common barriers to dermoscopy use cited by PCPs in the US include the cost of the dermatoscope, the time required to acquire proficiency, and the lack of financial reimbursement.16 True utilization of dermoscopy among PCPs is higher than this figure suggests due to the number of PCPs who access dermoscopic evaluations via teledermatology. All 21 Veterans Integrated Services Networks of the Veterans Health Administration (VHA) system, for instance, participate in teledermatology and jointly employ more than 1,150 trained telehealth clinical technicians who created a collective 107,000 teledermatology encounters with and without dermoscopy for evaluation by dermatologists in the most recent fiscal year(Martin Weinstock, written communication, October 2017). Nonetheless, it is necessary to determine the contribution that wider utilization of dermoscopy among PCPs would have on melanoma surveillance.

Studies show that dermoscopic algorithms improve the sensitivity while slightly decreasing the specificity of PCPs to detect melanoma compared with that of the naked eye examination. Dolianitis and colleagues demonstrated that a baseline sensitivity of 60.9% for melanoma detection improved to 85.4% with the 7-point checklist, 85.4% with Menzies method, and 77.5% with the ABCD rule, while the baseline specificity of 85.4% moderated to 73.0%, 77.7%, and 80.4%, respectively, among 61 medical practitioners after studying dermoscopy techniques from 2 CDs.21 Westerhoff and colleagues performed a randomized controlled trial with 74 PCPs to determine the effect of a minimal intervention on melanoma diagnostic accuracy. The intervention consisted of providing participants in the experimental group with an atlas of microscopic features common to melanoma to be read at the participants’ leisure, a 1-hour presentation on microscopy, and a 25-questionpractice quiz. Participants randomized to the intervention group improved their diagnostic accuracy from 57.8% to 75.9% with the use of dermoscopy. This group also experiencedimproved accuracy in its clinical diagnosis of melanoma from 54.6% to 62.7%.22

Argenziano and colleagues demonstrated similar results after PCPs attended a 4-hour workshop on dermoscopy. The 73 PCPs in this study evaluated 2,522 lesions randomized to naked eye examination or dermoscopy. The BMR, calculated from the data provided, improved from 12.6:1 to 10.5:1, respectively, when dermoscopy was incorporated into lesion analysis, while the sensitivity increased from 54.1% to 79.2% and the negative predictive value increased from 95.8% to 98.1%. It is important to note that the BMR and negative predictive value improved in tandem, indicating that PCPs were more discriminatory with their referrals for evaluation by dermatology while capturing a greater percentage of melanomas.23

These studies are not without limitations that preclude broad generalizations. For example, Dolianitis and colleagues and Westerhoff and colleagues provided participants with dermoscopic images of the lesions to be evaluated instead of requiring personal use of a dermatoscope, whereas the study by Argenziano and colleagues incorporated only 6 histopathologically proven malignant melanomas into each of the naked eye examination and the experimental dermoscopy groups.21-23 Yet these studies suggest that broader use of dermoscopy among PCPs could improve the accuracy of melanoma detection given clinically relevant training.

Several additional studies identify positive correlations associated with dermoscopy use among PCPs. A recent survey of 425 French general practitioners found that 8% of the study participants acknowledged owning a dermatoscope. Dermatoscope owners spent a statistically significant longer time analyzing each pigmented skin lesions, exhibited greater confidence in their analysis of pigmented lesions, and issued fewer overall referrals to dermatologists.24

Similarly, Rosendahl and colleagues evaluated the number needed to treat (NNT) (equivalent to the BMR) among 193 Australian PCPs and found that the NNT was inversely correlated to the frequency with which the physicians used dermoscopy. However, it was difficult to determine the definitive cause of the reduced NNT in this study because a similar effect was observed when NNT was evaluated based on general practitioner subspecialization.25 Again, despite limitations, these studies suggest that increased dermoscopy use among PCPs could reduce the morbidity of lifelong scarring as well as the short-term anxiety associated with a possible melanoma diagnosis.

 

 

Limitations

Even in the hands of a trained dermatologist, dermoscopy has limitations. Featureless melanoma is a term applied to melanoma lesions bereft of classical findings on both naked eye examination and dermoscopy. Menzies, a dermatologic pioneer in dermoscopy, acknowledged this limitation in 1996 while showing that 8% of melanomas evaded dermoscopic detection. He proceeded to discuss the importance of clinical history in melanoma detection because all of the featureless melanomas exhibited recent changes in size, shape, and/or color.26 More recently, sequential dermoscopy (mole mapping) imaging has been implemented to successfully identify these lesions.27 Thus, dermoscopy cannot replace dermatologists trained in the art of visual assessment with honed clinical diagnostic acumen. Rather, dermoscopy is a tool to enhance the assessment of clinically suspicious lesions and aid diagnostic discrimination of uncertain pigmented lesions.

Conclusion

Primary care physicians are on the frontline of medicine and often the first to have the opportunity to detect the presence of melanoma. Notably, 52.2% of the 884.7 million medical office visits performed annually in the US are with PCPs.28 Despite the benefits, dermoscopy is not uniformly used by dermatologists in the US. Of dermatologists practicing for more than 20 years, 76.2% use dermoscopy compared with 97.8% of dermatologists in practice for less than 5 years. This illustrates an increased use in tandem with dermatology residencies integrating dermoscopy training as a component of the curriculum, showing the importance of incorporating dermoscopy into medical school and residency training for PCPs..29-31 Guidelines regarding dermoscopy training and dermoscopic evaluation algorithms should be established, routinely taught in medical education, and actively incorporated into training curriculum for PCPs in order to improve patient care and realize the potential health care savings associated with the early diagnosis and treatment of melanoma. Dermoscopic-teledermatology consultations present a viable opportunity within the VHA to expedite access to care for veterans and simultaneously offer evaluative feedback on lesions to referring PCPs, as skilled, dermoscopy-trained dermatologists render the diagnoses. Given the devastating mortality rate of melanoma, continued multidisciplinary education on identifying melanoma is of the utmost importance for patient care. Widespread implementation of dermoscopy and dermoscopic-teledermatology consultations could save lives and slow the ever-increasing economic burden associated with melanoma treatment, costing $1.467 billion in 2016.32

References

1. Guy GP Jr, Thomas CC, Thompson T, Watson M, Massetti GM, Richardson LC. Vital signs: melanoma incidence and mortality trends and projections-United States, 1982-2030. MMWR Morb Mortal Wkly Rep. 2015;64(21):591-596.

2. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J Clin. 2018;68(1):7-30.

3. American Cancer Society. Cancer facts & figures 2017. Atlanta: American Cancer Society; 2017. https://www.cancer.org/content/dam/cancer-org/research/cancer-facts-and-statistics/annual-cancer-facts-and-figures/2017/cancer-facts-and-figures-2017.pdf. Accessed April 19, 2018.

4. Lea CS, Efird JT, Toland AE, Lewis DR, Phillips CJ. Melanoma incidence rates in active duty military personnel compared with a population-based registry in the United States, 2000-2007. Mil Med. 2014;179(3):247-253.

5. Thomas L, Puig S. Dermoscopy, digital dermoscopy and other diagnostic tools in the early detection of melanoma and follow-up of high-risk skin cancer patients. Acta Derm Venereol. 2017;97(218):14-21.

6. Marghoob AA, Usatine RP, Jaimes N. Dermoscopy for the family physician. Am Fam Physician. 2013;88(7):441-450.

7. Bafounta ML, Beauchet A, Aegerter P, Saiag P. Is dermoscopy (epiluminescence microscopy) useful for the diagnosis of melanoma? Results of a meta-analysis using techniques adapted to the evaluation of diagnostic tests. Arch Dermatol. 2001;137(10):1343-1350.

8. Terushkin V, Warycha M, Levy M, Kopf AW, Cohen DE, Polsky D. Analysis of the benign to malignant ratio of lesions biopsied by a general dermatologist before and after the adoption of dermoscopy. Arch Dermatol. 2010;146(3):343-344.

9. Wolner ZJ, Yélamos O, Liopyris K, Rogers T, Marchetti MA, Marghoob AA. Enhancing skin cancer diagnosis with dermoscopy. Dermatol Clin. 2017;35(4):417-437.

10. Carli P, Quercioli E, Sestini S, et al. Pattern analysis, not simplified algorithms, is the most reliable method for teaching dermoscopy for melanoma diagnosis to residents in dermatology. Br J Dermatol. 2003;148(5):981-984.

11. Argenziano G, Fabbrocini G, Carli P, De Giorgi V, Sammarco E, Delfino M. Epiluminescence microscopy for the diagnosis of doubtful melanocytic skin lesions. Comparison of the ABCD rule of dermatoscopy and a new 7-point checklist based on pattern analysis. Arch Dermatol. 1998;134(12):1563-1570.

12. Menzies SW, Ingvar C, Crotty KA, McCarthy WH. Frequency and morphologic characteristics of invasive melanomas lacking specific surface microscopic features. Arch Dermatol. 1996;132(10):1178-1182.

13. Nachbar F, Stolz W, Merkle T, et al. The ABCD rule of dermatoscopy. High prospective value in the diagnosis of doubtful melanocytic skin lesions. J Am Acad Dermatol. 1994;30(4):551-559.

14. Henning JS, Dusza SW, Wang SQ, et al. The CASH (color, architecture, symmetry, and homogeneity) algorithm for dermoscopy. J Am Acad Dermatol. 2007;56(1):45-52.

15. Rogers T, Marino M, Dusza SW, Bajaj S, Marchetti MA, Marghoob A. Triage amalgamated dermoscopic algorithm (TADA) for skin cancer screening. Dermatol Pract Concept. 2017;7(2):39-46.

16. Ackerman AB. No one should die of malignant melanoma. J Am Acad Dermatol. 1985;12(1):115-116.

17. Gandini S, Sera F, Cattaruzza MS, et al. Meta-analysis of risk factors for cutaneous melanoma: II: sun exposure. Eur J Cancer. 2005;41(1):45-60.

18. Gerbert B, Maurer T, Berger T, et al. Primary care physicians as gatekeepers in managed care. Primary care physicians’ and dermatologists’ skills at secondary prevention of skin cancer. Arch Dermatol. 1996;132(9):1030-1038.

19. Corbo MD, Wismer J. Agreement between dermatologists and primary care practitioners in the diagnosis of malignant melanoma: review of the literature. J Cutan Med Surg. 2012;16(5):306-310.

20. Morris JB, Alfonso SV, Hernandez N, Fernández MI. Examining the factors associated with past and present dermoscopy use among family physicians. Dermatol Pract Concept. 2017;7(4):63-70.

21. Dolianitis C, Kelly J, Wolfe R, Simpson P. Comparative performance of 4 dermoscopic algorithms by nonexperts for the diagnosis of melanocytic lesions. Arch Dermatol. 2005;141(8):1008-1014.

22. Westerhoff K, Mccarthy WH, Menzies SW. Increase in the sensitivity for melanoma diagnosis by primary care physicians using skin surface microscopy. Br J Dermatol. 2000;143(5):1016-1020.

23. Argenziano G, Puig S, Zalaudek I, et al. Dermoscopy improves accuracy of primary care physicians to triage lesions suggestive of skin cancer. J Clin Oncol. 2006;24(12):1877-1882.

24. Chappuis P, Duru G, Marchal O, Girier P, Dalle S, Thomas L. Dermoscopy, a useful tool for general practitioners in melanoma screening: a nationwide survey. Br J Dermatol. 2016;175(4):744-750.

25. Rosendahl C, Williams G, Eley D, et al. The impact of subspecialization and dermatoscopy use on accuracy of melanoma diagnosis among primary care doctors in Australia. J Am Acad Dermatol. 2012;67(5):846-852.

26. Menzies SW, Ingvar C, Crotty KA, McCarthy WH. Frequency and morphologic characteristics of invasive melanomas lacking specific surface microscopic features. Arch Dermatol. 1996;132(10):1178-1182.

27. Kittler H, Guitera P, Riedl E, et al. Identification of clinically featureless incipient melanoma using sequential dermoscopy imaging. Arch Dermatol. 2006;142(9):1113-1119.

28. Centers for Disease Control and Prevention. Ambulatory care use and physician office visits. https://www.cdc.gov/nchs/fastats/physician-visits.htm. Updated May 3, 2017. Accessed April 10, 2018.

29. Murzaku EC, Hayan S, Rao BK. Methods and rates of dermoscopy usage: a cross-sectional survey of US dermatologists stratified by years in practice. J Am Acad Dermatol. 2014;71(2):393-395.

30. Nehal KS, Oliveria SA, Marghoob AA, et al. Use of and beliefs about dermoscopy in the management of patients with pigmented lesions: a survey of dermatology residency programmes in the United States. Melanoma Res. 2002;12(6):601-605.

31. Wu TP, Newlove T, Smith L, Vuong CH, Stein JA, Polsky D. The importance of dedicated dermoscopy training during residency: a survey of US dermatology chief residents. J Am Acad Dermatol. 2013;68(6):1000-1005.

32. Lim HW, Collins SAB, Resneck JS Jr, et al. The burden of skin disease in the United States. J Am Acad Dermatol. 2017;76(5):958-972

References

1. Guy GP Jr, Thomas CC, Thompson T, Watson M, Massetti GM, Richardson LC. Vital signs: melanoma incidence and mortality trends and projections-United States, 1982-2030. MMWR Morb Mortal Wkly Rep. 2015;64(21):591-596.

2. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J Clin. 2018;68(1):7-30.

3. American Cancer Society. Cancer facts & figures 2017. Atlanta: American Cancer Society; 2017. https://www.cancer.org/content/dam/cancer-org/research/cancer-facts-and-statistics/annual-cancer-facts-and-figures/2017/cancer-facts-and-figures-2017.pdf. Accessed April 19, 2018.

4. Lea CS, Efird JT, Toland AE, Lewis DR, Phillips CJ. Melanoma incidence rates in active duty military personnel compared with a population-based registry in the United States, 2000-2007. Mil Med. 2014;179(3):247-253.

5. Thomas L, Puig S. Dermoscopy, digital dermoscopy and other diagnostic tools in the early detection of melanoma and follow-up of high-risk skin cancer patients. Acta Derm Venereol. 2017;97(218):14-21.

6. Marghoob AA, Usatine RP, Jaimes N. Dermoscopy for the family physician. Am Fam Physician. 2013;88(7):441-450.

7. Bafounta ML, Beauchet A, Aegerter P, Saiag P. Is dermoscopy (epiluminescence microscopy) useful for the diagnosis of melanoma? Results of a meta-analysis using techniques adapted to the evaluation of diagnostic tests. Arch Dermatol. 2001;137(10):1343-1350.

8. Terushkin V, Warycha M, Levy M, Kopf AW, Cohen DE, Polsky D. Analysis of the benign to malignant ratio of lesions biopsied by a general dermatologist before and after the adoption of dermoscopy. Arch Dermatol. 2010;146(3):343-344.

9. Wolner ZJ, Yélamos O, Liopyris K, Rogers T, Marchetti MA, Marghoob AA. Enhancing skin cancer diagnosis with dermoscopy. Dermatol Clin. 2017;35(4):417-437.

10. Carli P, Quercioli E, Sestini S, et al. Pattern analysis, not simplified algorithms, is the most reliable method for teaching dermoscopy for melanoma diagnosis to residents in dermatology. Br J Dermatol. 2003;148(5):981-984.

11. Argenziano G, Fabbrocini G, Carli P, De Giorgi V, Sammarco E, Delfino M. Epiluminescence microscopy for the diagnosis of doubtful melanocytic skin lesions. Comparison of the ABCD rule of dermatoscopy and a new 7-point checklist based on pattern analysis. Arch Dermatol. 1998;134(12):1563-1570.

12. Menzies SW, Ingvar C, Crotty KA, McCarthy WH. Frequency and morphologic characteristics of invasive melanomas lacking specific surface microscopic features. Arch Dermatol. 1996;132(10):1178-1182.

13. Nachbar F, Stolz W, Merkle T, et al. The ABCD rule of dermatoscopy. High prospective value in the diagnosis of doubtful melanocytic skin lesions. J Am Acad Dermatol. 1994;30(4):551-559.

14. Henning JS, Dusza SW, Wang SQ, et al. The CASH (color, architecture, symmetry, and homogeneity) algorithm for dermoscopy. J Am Acad Dermatol. 2007;56(1):45-52.

15. Rogers T, Marino M, Dusza SW, Bajaj S, Marchetti MA, Marghoob A. Triage amalgamated dermoscopic algorithm (TADA) for skin cancer screening. Dermatol Pract Concept. 2017;7(2):39-46.

16. Ackerman AB. No one should die of malignant melanoma. J Am Acad Dermatol. 1985;12(1):115-116.

17. Gandini S, Sera F, Cattaruzza MS, et al. Meta-analysis of risk factors for cutaneous melanoma: II: sun exposure. Eur J Cancer. 2005;41(1):45-60.

18. Gerbert B, Maurer T, Berger T, et al. Primary care physicians as gatekeepers in managed care. Primary care physicians’ and dermatologists’ skills at secondary prevention of skin cancer. Arch Dermatol. 1996;132(9):1030-1038.

19. Corbo MD, Wismer J. Agreement between dermatologists and primary care practitioners in the diagnosis of malignant melanoma: review of the literature. J Cutan Med Surg. 2012;16(5):306-310.

20. Morris JB, Alfonso SV, Hernandez N, Fernández MI. Examining the factors associated with past and present dermoscopy use among family physicians. Dermatol Pract Concept. 2017;7(4):63-70.

21. Dolianitis C, Kelly J, Wolfe R, Simpson P. Comparative performance of 4 dermoscopic algorithms by nonexperts for the diagnosis of melanocytic lesions. Arch Dermatol. 2005;141(8):1008-1014.

22. Westerhoff K, Mccarthy WH, Menzies SW. Increase in the sensitivity for melanoma diagnosis by primary care physicians using skin surface microscopy. Br J Dermatol. 2000;143(5):1016-1020.

23. Argenziano G, Puig S, Zalaudek I, et al. Dermoscopy improves accuracy of primary care physicians to triage lesions suggestive of skin cancer. J Clin Oncol. 2006;24(12):1877-1882.

24. Chappuis P, Duru G, Marchal O, Girier P, Dalle S, Thomas L. Dermoscopy, a useful tool for general practitioners in melanoma screening: a nationwide survey. Br J Dermatol. 2016;175(4):744-750.

25. Rosendahl C, Williams G, Eley D, et al. The impact of subspecialization and dermatoscopy use on accuracy of melanoma diagnosis among primary care doctors in Australia. J Am Acad Dermatol. 2012;67(5):846-852.

26. Menzies SW, Ingvar C, Crotty KA, McCarthy WH. Frequency and morphologic characteristics of invasive melanomas lacking specific surface microscopic features. Arch Dermatol. 1996;132(10):1178-1182.

27. Kittler H, Guitera P, Riedl E, et al. Identification of clinically featureless incipient melanoma using sequential dermoscopy imaging. Arch Dermatol. 2006;142(9):1113-1119.

28. Centers for Disease Control and Prevention. Ambulatory care use and physician office visits. https://www.cdc.gov/nchs/fastats/physician-visits.htm. Updated May 3, 2017. Accessed April 10, 2018.

29. Murzaku EC, Hayan S, Rao BK. Methods and rates of dermoscopy usage: a cross-sectional survey of US dermatologists stratified by years in practice. J Am Acad Dermatol. 2014;71(2):393-395.

30. Nehal KS, Oliveria SA, Marghoob AA, et al. Use of and beliefs about dermoscopy in the management of patients with pigmented lesions: a survey of dermatology residency programmes in the United States. Melanoma Res. 2002;12(6):601-605.

31. Wu TP, Newlove T, Smith L, Vuong CH, Stein JA, Polsky D. The importance of dedicated dermoscopy training during residency: a survey of US dermatology chief residents. J Am Acad Dermatol. 2013;68(6):1000-1005.

32. Lim HW, Collins SAB, Resneck JS Jr, et al. The burden of skin disease in the United States. J Am Acad Dermatol. 2017;76(5):958-972

Issue
Federal Practitioner - 35(4)s
Issue
Federal Practitioner - 35(4)s
Page Number
S39-S45
Page Number
S39-S45
Publications
Publications
Topics
Article Type
Display Headline
Using Dermoscopy to Identify Melanoma and Improve Diagnostic Discrimination
Display Headline
Using Dermoscopy to Identify Melanoma and Improve Diagnostic Discrimination
Sections
Citation Override
Fed Pract. 2018 May;35(suppl 4):S39-S45
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Article PDF Media

Pediatric cancers are on the rise

Article Type
Changed
Fri, 01/18/2019 - 17:39

 

– The incidence of many pediatric cancers are on the rise, and the increase is occurring in nearly all demographic groups studied, according to the latest data from the U.S. Centers for Disease Control and Prevention.

Pediatric cancers that increased significantly in incidence from 2001 through 2014, compared with previous time periods, include thyroid carcinoma, hepatic tumors, lymphomas, renal tumors, and brain tumors. Other cancer types remained unchanged, except malignant melanoma, which saw a significant decline in incidence over the same period, reported David A. Siegel, MD, of the Epidemic Intelligence Service at the CDC in Atlanta.

Neil Osterweil/MDedge News
Dr. David A. Siegel
“Some of the possible causes might be as benign as just increased ascertainment, but it might be more complicated, such as environmental exposures or population-based changes,” he said during a poster discussion session at the annual meeting of the American Society of Pediatric Hematology/Oncology.

Recent studies of trends in pediatric cancer have either used data from before 2010 or covered less than a third of the U.S. population, the investigators noted.

To get a more accurate estimate of current trends, the investigators relied on the United States Cancer Statistics, which combines data from the Surveillance, Epidemiology, and End Results (SEER) program and the National Program of Cancer Registries. Together, the combined databases cover 100% of the U.S. population.

Dr. Siegel and his colleagues looked at cancer incidence rates and trends among individuals younger than 20 years of age from across 48 states from 2001 to 2014 – Mississippi, Nevada, and the District of Columbia were not included.

They used a joinpoint regression method to calculate average annual percent change (AAPC) in rates, then stratified rates and trends by sex, age, and race/ethnicity; location; economic status; and cancer type.

 

 


During the 14-year period of the study, there were a total of 196,200 incident cases of pediatric cancer, for an overall cancer incidence rate of 173 per million. The pediatric cancer with the highest incident rate was leukemia of any type (45.6 per million), brain tumors (30.8), and lymphomas (26.0).

Incidence rates were highest among males, patients from infancy through age 4, non-Hispanic whites, children who live in the Northeast region, those who live in the wealthiest counties, and those who live in urban/metropolitan counties. The overall pediatric cancer incidence rate increased, with an AAPC of 0.7 (95% confidence interval, 0.5-0.8).

“Rates increased in each stratum of sex, age, and race/ethnicity (except non-Hispanic American Indian/Alaska Native), region, economic status, and rural/urban classification,” the investigators wrote.

Cancers with significantly increased AAPC included thyroid carcinomas (AAPC, 4.8), hepatic tumors (2.5), lymphomas (1.7), renal tumors (0.6), and brain tumors (all types, 0.4).
 

 


There were no significant changes in the incidence of either germ cell cancer, retinoblastoma, leukemia, neuroblastoma, soft-tissue sarcomas, or bone tumors.

The only significant decrease over the study period was in the incidence of melanoma in children (–2.6).

“Possible causes of increasing rates might include changes in diagnostic, coding, and reporting standards, increased detection, population-based changes (such as increasing obesity), and environmental exposures,” they wrote.

Public health campaigns about the dangers of UV exposure and promoting the use of sunscreens may account for the decline in the incidence of malignant melanoma, they suggested.

The study was supported by the CDC. Dr. Siegel and coauthors are CDC employees. They reported having no conflicts of interest.

SOURCE: Siegel DA et al. ASPHO 2018, Abstract 605.

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

 

– The incidence of many pediatric cancers are on the rise, and the increase is occurring in nearly all demographic groups studied, according to the latest data from the U.S. Centers for Disease Control and Prevention.

Pediatric cancers that increased significantly in incidence from 2001 through 2014, compared with previous time periods, include thyroid carcinoma, hepatic tumors, lymphomas, renal tumors, and brain tumors. Other cancer types remained unchanged, except malignant melanoma, which saw a significant decline in incidence over the same period, reported David A. Siegel, MD, of the Epidemic Intelligence Service at the CDC in Atlanta.

Neil Osterweil/MDedge News
Dr. David A. Siegel
“Some of the possible causes might be as benign as just increased ascertainment, but it might be more complicated, such as environmental exposures or population-based changes,” he said during a poster discussion session at the annual meeting of the American Society of Pediatric Hematology/Oncology.

Recent studies of trends in pediatric cancer have either used data from before 2010 or covered less than a third of the U.S. population, the investigators noted.

To get a more accurate estimate of current trends, the investigators relied on the United States Cancer Statistics, which combines data from the Surveillance, Epidemiology, and End Results (SEER) program and the National Program of Cancer Registries. Together, the combined databases cover 100% of the U.S. population.

Dr. Siegel and his colleagues looked at cancer incidence rates and trends among individuals younger than 20 years of age from across 48 states from 2001 to 2014 – Mississippi, Nevada, and the District of Columbia were not included.

They used a joinpoint regression method to calculate average annual percent change (AAPC) in rates, then stratified rates and trends by sex, age, and race/ethnicity; location; economic status; and cancer type.

 

 


During the 14-year period of the study, there were a total of 196,200 incident cases of pediatric cancer, for an overall cancer incidence rate of 173 per million. The pediatric cancer with the highest incident rate was leukemia of any type (45.6 per million), brain tumors (30.8), and lymphomas (26.0).

Incidence rates were highest among males, patients from infancy through age 4, non-Hispanic whites, children who live in the Northeast region, those who live in the wealthiest counties, and those who live in urban/metropolitan counties. The overall pediatric cancer incidence rate increased, with an AAPC of 0.7 (95% confidence interval, 0.5-0.8).

“Rates increased in each stratum of sex, age, and race/ethnicity (except non-Hispanic American Indian/Alaska Native), region, economic status, and rural/urban classification,” the investigators wrote.

Cancers with significantly increased AAPC included thyroid carcinomas (AAPC, 4.8), hepatic tumors (2.5), lymphomas (1.7), renal tumors (0.6), and brain tumors (all types, 0.4).
 

 


There were no significant changes in the incidence of either germ cell cancer, retinoblastoma, leukemia, neuroblastoma, soft-tissue sarcomas, or bone tumors.

The only significant decrease over the study period was in the incidence of melanoma in children (–2.6).

“Possible causes of increasing rates might include changes in diagnostic, coding, and reporting standards, increased detection, population-based changes (such as increasing obesity), and environmental exposures,” they wrote.

Public health campaigns about the dangers of UV exposure and promoting the use of sunscreens may account for the decline in the incidence of malignant melanoma, they suggested.

The study was supported by the CDC. Dr. Siegel and coauthors are CDC employees. They reported having no conflicts of interest.

SOURCE: Siegel DA et al. ASPHO 2018, Abstract 605.

 

– The incidence of many pediatric cancers are on the rise, and the increase is occurring in nearly all demographic groups studied, according to the latest data from the U.S. Centers for Disease Control and Prevention.

Pediatric cancers that increased significantly in incidence from 2001 through 2014, compared with previous time periods, include thyroid carcinoma, hepatic tumors, lymphomas, renal tumors, and brain tumors. Other cancer types remained unchanged, except malignant melanoma, which saw a significant decline in incidence over the same period, reported David A. Siegel, MD, of the Epidemic Intelligence Service at the CDC in Atlanta.

Neil Osterweil/MDedge News
Dr. David A. Siegel
“Some of the possible causes might be as benign as just increased ascertainment, but it might be more complicated, such as environmental exposures or population-based changes,” he said during a poster discussion session at the annual meeting of the American Society of Pediatric Hematology/Oncology.

Recent studies of trends in pediatric cancer have either used data from before 2010 or covered less than a third of the U.S. population, the investigators noted.

To get a more accurate estimate of current trends, the investigators relied on the United States Cancer Statistics, which combines data from the Surveillance, Epidemiology, and End Results (SEER) program and the National Program of Cancer Registries. Together, the combined databases cover 100% of the U.S. population.

Dr. Siegel and his colleagues looked at cancer incidence rates and trends among individuals younger than 20 years of age from across 48 states from 2001 to 2014 – Mississippi, Nevada, and the District of Columbia were not included.

They used a joinpoint regression method to calculate average annual percent change (AAPC) in rates, then stratified rates and trends by sex, age, and race/ethnicity; location; economic status; and cancer type.

 

 


During the 14-year period of the study, there were a total of 196,200 incident cases of pediatric cancer, for an overall cancer incidence rate of 173 per million. The pediatric cancer with the highest incident rate was leukemia of any type (45.6 per million), brain tumors (30.8), and lymphomas (26.0).

Incidence rates were highest among males, patients from infancy through age 4, non-Hispanic whites, children who live in the Northeast region, those who live in the wealthiest counties, and those who live in urban/metropolitan counties. The overall pediatric cancer incidence rate increased, with an AAPC of 0.7 (95% confidence interval, 0.5-0.8).

“Rates increased in each stratum of sex, age, and race/ethnicity (except non-Hispanic American Indian/Alaska Native), region, economic status, and rural/urban classification,” the investigators wrote.

Cancers with significantly increased AAPC included thyroid carcinomas (AAPC, 4.8), hepatic tumors (2.5), lymphomas (1.7), renal tumors (0.6), and brain tumors (all types, 0.4).
 

 


There were no significant changes in the incidence of either germ cell cancer, retinoblastoma, leukemia, neuroblastoma, soft-tissue sarcomas, or bone tumors.

The only significant decrease over the study period was in the incidence of melanoma in children (–2.6).

“Possible causes of increasing rates might include changes in diagnostic, coding, and reporting standards, increased detection, population-based changes (such as increasing obesity), and environmental exposures,” they wrote.

Public health campaigns about the dangers of UV exposure and promoting the use of sunscreens may account for the decline in the incidence of malignant melanoma, they suggested.

The study was supported by the CDC. Dr. Siegel and coauthors are CDC employees. They reported having no conflicts of interest.

SOURCE: Siegel DA et al. ASPHO 2018, Abstract 605.

Publications
Publications
Topics
Article Type
Sections
Article Source

REPORTING FROM ASPHO 2018

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

 

Key clinical point: Pediatric cancer incidence in the United States has increased significantly in recent decades.Major finding: From 2001 to 2014, there were 196,200 incident cases of pediatric cancer for an overall cancer incidence rate of 173 per 1 million.

Study details: A review of data from the United States Cancer Statistics for children under age 20.

Disclosures: The CDC supported the study. Dr. Siegel and his coauthors are CDC employees. They reported having no conflicts of interest.

Source: Siegel DA et al. ASPHO 2018, Abstract 605.

Disqus Comments
Default
Use ProPublica

Is cancer immunotherapy more effective in men than women?

Sex differences in cancer immunotherapy outcomes unclear
Article Type
Changed
Wed, 05/26/2021 - 13:49

 

Cancer immunotherapy using checkpoint inhibitors may achieve greater mortality reductions in men than they do in women, new research has suggested.

In a meta-analysis and systematic review published in Lancet Oncology, researchers analyzed 20 randomized, controlled trials of immune checkpoint inhibitors that included detail on overall survival and patients’ sex; altogether, these studies involved 11,351 patients with advanced or metastatic cancers.

They found that while men treated with checkpoint inhibitors had a significant 28% reduced risk of death, compared with male controls, the survival benefit in women was smaller (14% reduced risk of death, compared with female controls).

Fabio Conforti, MD, from the European Institute of Oncology, Milan, and coauthors commented that the magnitude of the difference between the effect seen men and that in women was clinically significant.

“The pooled reduction of risk of death was double the size for male patients than for female patients – a difference that is similar to the size of the difference in survival benefit observed between patients with non–small cell lung cancer with PD-L1 positive (greater than 1%) tumors versus negative tumors, who were treated with anti-PD-1,” they wrote.

This difference between the benefit seen men and that in women was evident across all the subgroups in the study, which included subgroups based on cancer histotype, line of treatment, drugs used, and type of control.

However there was greater heterogeneity in the magnitude of the effect of checkpoint inhibitors on mortality in men than there was in women. The authors suggested this could be explained by the fact that the drugs have lower efficacy in women and this may therefore reduce the variability of results when compared with those in men.

 

 


The authors also looked at whether the studies that compared immunotherapies with nonimmunological therapies might show a different effect, but they still found a significantly higher benefit in men, compared with women.

The overall study population was two-thirds male and one-third female. The checkpoint inhibitors used were ipilimumab, tremelimumab, nivolumab, and pembrolizumab, and the trials were conducted in patients with melanoma, non–small cell lung cancer, head and neck cancer, renal cell carcinoma, urothelial tumors, gastric tumors, and mesothelioma.

Men have almost double the risk of mortality from cancer than do women, the authors said, with the greatest differences seen in melanoma, lung cancer, larynx cancer, esophagus cancer, and bladder cancer.

“This male-biased mortality is hypothesized to reflect differences not only in behavioral and biological factors, including causes of cancer and hormonal regulation, but also in the immune system.”
 

 


Despite this, sex is rarely taken into account when new therapeutic approaches are tested, the authors said.

They also commented on the fact that there was a relatively low number of women included in each trial, an issue that was recognized as far back as the 1990s as a major problem in medical trials.

“Our results further highlight this problem, showing clinically relevant differences in the efficacy of two important classes of immunological drugs, namely anti–CTLA-4 and anti–PD-1 antibodies, when compared with controls in male and female patients with advanced solid tumors,” they wrote.

They noted that they couldn’t exclude the possibility that the effect may be the result of other variables that were distributed differently between the sexes. However, they also qualified this by saying that variables known to affect the efficacy of immune checkpoint inhibitors, such as PD-L1 expression and mutation status, were not likely to explain the results.
 

 


Given their findings, the authors said a patient’s sex should be taken into account when weighing the risks and benefits of checkpoint inhibitors given the magnitude of benefit was sex-dependent. They also called for future immunotherapy studies to include more women.

No funding or conflicts of interest were declared.

SOURCE: Conforti F et al. Lancet Oncol. 2018 May 16. doi: 10.1016/S1470-2045(18)30261-4.

Body

 

While cancer immunotherapy represents one of the most significant clinical advances in cancer treatment in the past decade, the basic but important clinical question about different effects between men and woman has not been addressed until now. The authors of this study are to be congratulated on such a comprehensive and well-conducted analysis, but the data does not completely support their final conclusion that checkpoint inhibitors benefit men more than women.

There are a large number of baseline characteristics of solid tumors that might differ between men and women and that have also been reported to impact the outcomes of patients treated with checkpoint inhibitors. Some of these may be lifestyle or behavioral characteristics – such as different smoking habits between men and women with non–small cell lung cancer – or differences in the distribution of oncogenic driver mutations between men and women.

We should therefore be cautious in jumping to conclusions and changing the current standard of care with respect to checkpoint inhibitors. In particular, we should not be denying treatment to women who are otherwise indicated for checkpoint inhibitors, based on these findings.

Omar Abdel-Rahman, MD, is from the clinical oncology department of the faculty of medicine at Ain Shams University in Cairo and from the Tom Baker Cancer Centre in Calgary. These comments are taken from an accompanying editorial (Lancet Oncol. 2018 May 16. doi: 10.1016/S1470-2045[18]30270-5.) No conflicts of interest were declared.

Publications
Topics
Sections
Body

 

While cancer immunotherapy represents one of the most significant clinical advances in cancer treatment in the past decade, the basic but important clinical question about different effects between men and woman has not been addressed until now. The authors of this study are to be congratulated on such a comprehensive and well-conducted analysis, but the data does not completely support their final conclusion that checkpoint inhibitors benefit men more than women.

There are a large number of baseline characteristics of solid tumors that might differ between men and women and that have also been reported to impact the outcomes of patients treated with checkpoint inhibitors. Some of these may be lifestyle or behavioral characteristics – such as different smoking habits between men and women with non–small cell lung cancer – or differences in the distribution of oncogenic driver mutations between men and women.

We should therefore be cautious in jumping to conclusions and changing the current standard of care with respect to checkpoint inhibitors. In particular, we should not be denying treatment to women who are otherwise indicated for checkpoint inhibitors, based on these findings.

Omar Abdel-Rahman, MD, is from the clinical oncology department of the faculty of medicine at Ain Shams University in Cairo and from the Tom Baker Cancer Centre in Calgary. These comments are taken from an accompanying editorial (Lancet Oncol. 2018 May 16. doi: 10.1016/S1470-2045[18]30270-5.) No conflicts of interest were declared.

Body

 

While cancer immunotherapy represents one of the most significant clinical advances in cancer treatment in the past decade, the basic but important clinical question about different effects between men and woman has not been addressed until now. The authors of this study are to be congratulated on such a comprehensive and well-conducted analysis, but the data does not completely support their final conclusion that checkpoint inhibitors benefit men more than women.

There are a large number of baseline characteristics of solid tumors that might differ between men and women and that have also been reported to impact the outcomes of patients treated with checkpoint inhibitors. Some of these may be lifestyle or behavioral characteristics – such as different smoking habits between men and women with non–small cell lung cancer – or differences in the distribution of oncogenic driver mutations between men and women.

We should therefore be cautious in jumping to conclusions and changing the current standard of care with respect to checkpoint inhibitors. In particular, we should not be denying treatment to women who are otherwise indicated for checkpoint inhibitors, based on these findings.

Omar Abdel-Rahman, MD, is from the clinical oncology department of the faculty of medicine at Ain Shams University in Cairo and from the Tom Baker Cancer Centre in Calgary. These comments are taken from an accompanying editorial (Lancet Oncol. 2018 May 16. doi: 10.1016/S1470-2045[18]30270-5.) No conflicts of interest were declared.

Title
Sex differences in cancer immunotherapy outcomes unclear
Sex differences in cancer immunotherapy outcomes unclear

 

Cancer immunotherapy using checkpoint inhibitors may achieve greater mortality reductions in men than they do in women, new research has suggested.

In a meta-analysis and systematic review published in Lancet Oncology, researchers analyzed 20 randomized, controlled trials of immune checkpoint inhibitors that included detail on overall survival and patients’ sex; altogether, these studies involved 11,351 patients with advanced or metastatic cancers.

They found that while men treated with checkpoint inhibitors had a significant 28% reduced risk of death, compared with male controls, the survival benefit in women was smaller (14% reduced risk of death, compared with female controls).

Fabio Conforti, MD, from the European Institute of Oncology, Milan, and coauthors commented that the magnitude of the difference between the effect seen men and that in women was clinically significant.

“The pooled reduction of risk of death was double the size for male patients than for female patients – a difference that is similar to the size of the difference in survival benefit observed between patients with non–small cell lung cancer with PD-L1 positive (greater than 1%) tumors versus negative tumors, who were treated with anti-PD-1,” they wrote.

This difference between the benefit seen men and that in women was evident across all the subgroups in the study, which included subgroups based on cancer histotype, line of treatment, drugs used, and type of control.

However there was greater heterogeneity in the magnitude of the effect of checkpoint inhibitors on mortality in men than there was in women. The authors suggested this could be explained by the fact that the drugs have lower efficacy in women and this may therefore reduce the variability of results when compared with those in men.

 

 


The authors also looked at whether the studies that compared immunotherapies with nonimmunological therapies might show a different effect, but they still found a significantly higher benefit in men, compared with women.

The overall study population was two-thirds male and one-third female. The checkpoint inhibitors used were ipilimumab, tremelimumab, nivolumab, and pembrolizumab, and the trials were conducted in patients with melanoma, non–small cell lung cancer, head and neck cancer, renal cell carcinoma, urothelial tumors, gastric tumors, and mesothelioma.

Men have almost double the risk of mortality from cancer than do women, the authors said, with the greatest differences seen in melanoma, lung cancer, larynx cancer, esophagus cancer, and bladder cancer.

“This male-biased mortality is hypothesized to reflect differences not only in behavioral and biological factors, including causes of cancer and hormonal regulation, but also in the immune system.”
 

 


Despite this, sex is rarely taken into account when new therapeutic approaches are tested, the authors said.

They also commented on the fact that there was a relatively low number of women included in each trial, an issue that was recognized as far back as the 1990s as a major problem in medical trials.

“Our results further highlight this problem, showing clinically relevant differences in the efficacy of two important classes of immunological drugs, namely anti–CTLA-4 and anti–PD-1 antibodies, when compared with controls in male and female patients with advanced solid tumors,” they wrote.

They noted that they couldn’t exclude the possibility that the effect may be the result of other variables that were distributed differently between the sexes. However, they also qualified this by saying that variables known to affect the efficacy of immune checkpoint inhibitors, such as PD-L1 expression and mutation status, were not likely to explain the results.
 

 


Given their findings, the authors said a patient’s sex should be taken into account when weighing the risks and benefits of checkpoint inhibitors given the magnitude of benefit was sex-dependent. They also called for future immunotherapy studies to include more women.

No funding or conflicts of interest were declared.

SOURCE: Conforti F et al. Lancet Oncol. 2018 May 16. doi: 10.1016/S1470-2045(18)30261-4.

 

Cancer immunotherapy using checkpoint inhibitors may achieve greater mortality reductions in men than they do in women, new research has suggested.

In a meta-analysis and systematic review published in Lancet Oncology, researchers analyzed 20 randomized, controlled trials of immune checkpoint inhibitors that included detail on overall survival and patients’ sex; altogether, these studies involved 11,351 patients with advanced or metastatic cancers.

They found that while men treated with checkpoint inhibitors had a significant 28% reduced risk of death, compared with male controls, the survival benefit in women was smaller (14% reduced risk of death, compared with female controls).

Fabio Conforti, MD, from the European Institute of Oncology, Milan, and coauthors commented that the magnitude of the difference between the effect seen men and that in women was clinically significant.

“The pooled reduction of risk of death was double the size for male patients than for female patients – a difference that is similar to the size of the difference in survival benefit observed between patients with non–small cell lung cancer with PD-L1 positive (greater than 1%) tumors versus negative tumors, who were treated with anti-PD-1,” they wrote.

This difference between the benefit seen men and that in women was evident across all the subgroups in the study, which included subgroups based on cancer histotype, line of treatment, drugs used, and type of control.

However there was greater heterogeneity in the magnitude of the effect of checkpoint inhibitors on mortality in men than there was in women. The authors suggested this could be explained by the fact that the drugs have lower efficacy in women and this may therefore reduce the variability of results when compared with those in men.

 

 


The authors also looked at whether the studies that compared immunotherapies with nonimmunological therapies might show a different effect, but they still found a significantly higher benefit in men, compared with women.

The overall study population was two-thirds male and one-third female. The checkpoint inhibitors used were ipilimumab, tremelimumab, nivolumab, and pembrolizumab, and the trials were conducted in patients with melanoma, non–small cell lung cancer, head and neck cancer, renal cell carcinoma, urothelial tumors, gastric tumors, and mesothelioma.

Men have almost double the risk of mortality from cancer than do women, the authors said, with the greatest differences seen in melanoma, lung cancer, larynx cancer, esophagus cancer, and bladder cancer.

“This male-biased mortality is hypothesized to reflect differences not only in behavioral and biological factors, including causes of cancer and hormonal regulation, but also in the immune system.”
 

 


Despite this, sex is rarely taken into account when new therapeutic approaches are tested, the authors said.

They also commented on the fact that there was a relatively low number of women included in each trial, an issue that was recognized as far back as the 1990s as a major problem in medical trials.

“Our results further highlight this problem, showing clinically relevant differences in the efficacy of two important classes of immunological drugs, namely anti–CTLA-4 and anti–PD-1 antibodies, when compared with controls in male and female patients with advanced solid tumors,” they wrote.

They noted that they couldn’t exclude the possibility that the effect may be the result of other variables that were distributed differently between the sexes. However, they also qualified this by saying that variables known to affect the efficacy of immune checkpoint inhibitors, such as PD-L1 expression and mutation status, were not likely to explain the results.
 

 


Given their findings, the authors said a patient’s sex should be taken into account when weighing the risks and benefits of checkpoint inhibitors given the magnitude of benefit was sex-dependent. They also called for future immunotherapy studies to include more women.

No funding or conflicts of interest were declared.

SOURCE: Conforti F et al. Lancet Oncol. 2018 May 16. doi: 10.1016/S1470-2045(18)30261-4.

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM LANCET ONCOLOGY

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

 

Key clinical point: Checkpoint inhibitors are linked with greater mortality reductions in men than in women.

Major finding: Checkpoint inhibitors are associated with a 28% reduction in cancer mortality in men and 14% in women.

Study details: Systematic review and meta-analysis of 20 randomized, controlled trials involving 11,351 patients.

Disclosures: No funding or conflicts of interest were declared.

Source: Conforti F et al. Lancet Oncol. 2018 May 16. doi: 10.1016/S1470-2045(18)30261-4.

Disqus Comments
Default
Use ProPublica

Mask provides effective, cheap protection from hazardous electrocautery plumes

Article Type
Changed
Mon, 01/14/2019 - 10:23

 

– Routine use of an N95 mask during electrocautery is an effective and inexpensive way for dermatologic surgeons to protect themselves from toxic, airborne particulate matter in the smoke generated during the procedure, Emily de Golian, MD, said at the annual meeting of the American College of Mohs Surgery.

“Our data suggest clear dominance of N95 masks at filtering electrocautery particulate matter over commonly utilized basic procedural masks, as well as superiority to the laser masks that are used in hair removal procedures and ablative procedures in cosmetic clinics,” commented Dr. de Golian, a Mohs micrographic surgery fellow at the University of California, San Diego.

Bruce Jancin/MDedge News
Dr. Emily de Golian
This is an inexpensive protective strategy, and Dr. de Golian priced N95 masks on Amazon at 94 cents each. While that’s more than the 11 cents per standard procedural mask with earloops or 83 cents for a laser mask on Amazon, it’s still small potatoes considering the enhanced protection she documented in her study. Of note, all of the masks cost more when purchased from medical supply companies.

This matter of self-protection from the effects of electrocautery smoke plumes deserves greater attention from the dermatologic community, according to Dr. de Golian. There is solid evidence that these plumes contain high concentrations of known carcinogens, including benzene, acetonitrile, and butadiene – indeed, concentrations far in excess of what’s found in second-hand cigarette smoke. Moreover, many of these airborne carcinogens and other toxins have been linked to leukemia, neurologic disorders, lung cancer, thrombotic disorders, lung disease, and infectious disease transmission, albeit not convincingly so to date in dermatologic surgeons. But why wait for definitive evidence to accrue?

“In light of these hazards – and according to governmental guidelines – dermatologic surgeons would be wise to adopt protective measures during surgical procedures,” Dr. de Golian said.

But they haven’t. She cited a national survey conducted several years ago by a colleague in which 79% of the 316 responding dermatologic surgeons indicated they use no smoke management whatsoever, neither masks nor a local exhaust evacuation system. Only 10% employed smoke management 25%-50% of the time during electrocautery, and a scant 11% of dermatologic surgeons did so at least 75% of the time (Dermatol Surg. 2014 Dec;40[12]:1373-7).

Given the far more substantial expense of installing an office smoke evacuation system, mask filtration becomes an attractive alternative. But the relative efficacy of the various types of masks in blocking fine and ultrafine particulate matter contained in electrocautery plumes hadn’t previously been systematically studied. This created the impetus for Dr. de Golian’s study.

 

 



ninuns/iStock/Getty Images Plus
She placed N95 masks, basic procedural masks, and laser masks over the probes of two commercially available airborne particle counters, one capturing ultrafine particles less than 1 mcm in size, the other for particles greater than 2.5 mcm. She sealed the mask edges to the probes with tape to eliminate loose fit as a confounder. Then she and her coinvestigators performed monopolar electrocautery on discarded surgical Burow triangle tissue specimens to order to create smoke plumes and test mask effectiveness. This process was repeated five times for each type of mask, as well as for a no-mask control condition.

The N95 masks were the clear winner, particularly when it came to filtering the ultrafine particles, which are of greatest concern because they remain suspended in air longer and penetrate deeper into the respiratory tract than larger particles. The N95 masks proved superior to procedural masks, which in turn were significantly more effective than the laser masks. The differences between mask performance for larger particle filtration were smaller, although the N95 remained number one. She noted that the study results probably underestimate the true filtration efficacy of N95 masks, since they form a tighter seal with the face in clinical practice than with the other two mask types.

Mask self-protection “is easily applicable in your own practice, and it meets NIOSH-recommended [National Institute for Occupational Safety and Health] standards for safety in the workplace,” the dermatologist noted.

In the next phase of her research, she plans to evaluate the optimal technology and techniques of smoke evacuation in the surgical suite. That’s an attractive method because it protects everyone in the room, not just the surgeon. And while the practitioner survey indicates this technology isn’t widely used by dermatologic surgeons on a routine basis at present, that could change, particularly in the current era in which patient-reported outcomes and satisfaction surveys have taken on added weight.

 

 


“Patients prefer not smelling their own tissue burning,” Dr. de Golian said.

She reported no financial conflicts regarding her study, which was conducted free of commercial support.
 
Meeting/Event
Publications
Topics
Sections
Meeting/Event
Meeting/Event

 

– Routine use of an N95 mask during electrocautery is an effective and inexpensive way for dermatologic surgeons to protect themselves from toxic, airborne particulate matter in the smoke generated during the procedure, Emily de Golian, MD, said at the annual meeting of the American College of Mohs Surgery.

“Our data suggest clear dominance of N95 masks at filtering electrocautery particulate matter over commonly utilized basic procedural masks, as well as superiority to the laser masks that are used in hair removal procedures and ablative procedures in cosmetic clinics,” commented Dr. de Golian, a Mohs micrographic surgery fellow at the University of California, San Diego.

Bruce Jancin/MDedge News
Dr. Emily de Golian
This is an inexpensive protective strategy, and Dr. de Golian priced N95 masks on Amazon at 94 cents each. While that’s more than the 11 cents per standard procedural mask with earloops or 83 cents for a laser mask on Amazon, it’s still small potatoes considering the enhanced protection she documented in her study. Of note, all of the masks cost more when purchased from medical supply companies.

This matter of self-protection from the effects of electrocautery smoke plumes deserves greater attention from the dermatologic community, according to Dr. de Golian. There is solid evidence that these plumes contain high concentrations of known carcinogens, including benzene, acetonitrile, and butadiene – indeed, concentrations far in excess of what’s found in second-hand cigarette smoke. Moreover, many of these airborne carcinogens and other toxins have been linked to leukemia, neurologic disorders, lung cancer, thrombotic disorders, lung disease, and infectious disease transmission, albeit not convincingly so to date in dermatologic surgeons. But why wait for definitive evidence to accrue?

“In light of these hazards – and according to governmental guidelines – dermatologic surgeons would be wise to adopt protective measures during surgical procedures,” Dr. de Golian said.

But they haven’t. She cited a national survey conducted several years ago by a colleague in which 79% of the 316 responding dermatologic surgeons indicated they use no smoke management whatsoever, neither masks nor a local exhaust evacuation system. Only 10% employed smoke management 25%-50% of the time during electrocautery, and a scant 11% of dermatologic surgeons did so at least 75% of the time (Dermatol Surg. 2014 Dec;40[12]:1373-7).

Given the far more substantial expense of installing an office smoke evacuation system, mask filtration becomes an attractive alternative. But the relative efficacy of the various types of masks in blocking fine and ultrafine particulate matter contained in electrocautery plumes hadn’t previously been systematically studied. This created the impetus for Dr. de Golian’s study.

 

 



ninuns/iStock/Getty Images Plus
She placed N95 masks, basic procedural masks, and laser masks over the probes of two commercially available airborne particle counters, one capturing ultrafine particles less than 1 mcm in size, the other for particles greater than 2.5 mcm. She sealed the mask edges to the probes with tape to eliminate loose fit as a confounder. Then she and her coinvestigators performed monopolar electrocautery on discarded surgical Burow triangle tissue specimens to order to create smoke plumes and test mask effectiveness. This process was repeated five times for each type of mask, as well as for a no-mask control condition.

The N95 masks were the clear winner, particularly when it came to filtering the ultrafine particles, which are of greatest concern because they remain suspended in air longer and penetrate deeper into the respiratory tract than larger particles. The N95 masks proved superior to procedural masks, which in turn were significantly more effective than the laser masks. The differences between mask performance for larger particle filtration were smaller, although the N95 remained number one. She noted that the study results probably underestimate the true filtration efficacy of N95 masks, since they form a tighter seal with the face in clinical practice than with the other two mask types.

Mask self-protection “is easily applicable in your own practice, and it meets NIOSH-recommended [National Institute for Occupational Safety and Health] standards for safety in the workplace,” the dermatologist noted.

In the next phase of her research, she plans to evaluate the optimal technology and techniques of smoke evacuation in the surgical suite. That’s an attractive method because it protects everyone in the room, not just the surgeon. And while the practitioner survey indicates this technology isn’t widely used by dermatologic surgeons on a routine basis at present, that could change, particularly in the current era in which patient-reported outcomes and satisfaction surveys have taken on added weight.

 

 


“Patients prefer not smelling their own tissue burning,” Dr. de Golian said.

She reported no financial conflicts regarding her study, which was conducted free of commercial support.
 

 

– Routine use of an N95 mask during electrocautery is an effective and inexpensive way for dermatologic surgeons to protect themselves from toxic, airborne particulate matter in the smoke generated during the procedure, Emily de Golian, MD, said at the annual meeting of the American College of Mohs Surgery.

“Our data suggest clear dominance of N95 masks at filtering electrocautery particulate matter over commonly utilized basic procedural masks, as well as superiority to the laser masks that are used in hair removal procedures and ablative procedures in cosmetic clinics,” commented Dr. de Golian, a Mohs micrographic surgery fellow at the University of California, San Diego.

Bruce Jancin/MDedge News
Dr. Emily de Golian
This is an inexpensive protective strategy, and Dr. de Golian priced N95 masks on Amazon at 94 cents each. While that’s more than the 11 cents per standard procedural mask with earloops or 83 cents for a laser mask on Amazon, it’s still small potatoes considering the enhanced protection she documented in her study. Of note, all of the masks cost more when purchased from medical supply companies.

This matter of self-protection from the effects of electrocautery smoke plumes deserves greater attention from the dermatologic community, according to Dr. de Golian. There is solid evidence that these plumes contain high concentrations of known carcinogens, including benzene, acetonitrile, and butadiene – indeed, concentrations far in excess of what’s found in second-hand cigarette smoke. Moreover, many of these airborne carcinogens and other toxins have been linked to leukemia, neurologic disorders, lung cancer, thrombotic disorders, lung disease, and infectious disease transmission, albeit not convincingly so to date in dermatologic surgeons. But why wait for definitive evidence to accrue?

“In light of these hazards – and according to governmental guidelines – dermatologic surgeons would be wise to adopt protective measures during surgical procedures,” Dr. de Golian said.

But they haven’t. She cited a national survey conducted several years ago by a colleague in which 79% of the 316 responding dermatologic surgeons indicated they use no smoke management whatsoever, neither masks nor a local exhaust evacuation system. Only 10% employed smoke management 25%-50% of the time during electrocautery, and a scant 11% of dermatologic surgeons did so at least 75% of the time (Dermatol Surg. 2014 Dec;40[12]:1373-7).

Given the far more substantial expense of installing an office smoke evacuation system, mask filtration becomes an attractive alternative. But the relative efficacy of the various types of masks in blocking fine and ultrafine particulate matter contained in electrocautery plumes hadn’t previously been systematically studied. This created the impetus for Dr. de Golian’s study.

 

 



ninuns/iStock/Getty Images Plus
She placed N95 masks, basic procedural masks, and laser masks over the probes of two commercially available airborne particle counters, one capturing ultrafine particles less than 1 mcm in size, the other for particles greater than 2.5 mcm. She sealed the mask edges to the probes with tape to eliminate loose fit as a confounder. Then she and her coinvestigators performed monopolar electrocautery on discarded surgical Burow triangle tissue specimens to order to create smoke plumes and test mask effectiveness. This process was repeated five times for each type of mask, as well as for a no-mask control condition.

The N95 masks were the clear winner, particularly when it came to filtering the ultrafine particles, which are of greatest concern because they remain suspended in air longer and penetrate deeper into the respiratory tract than larger particles. The N95 masks proved superior to procedural masks, which in turn were significantly more effective than the laser masks. The differences between mask performance for larger particle filtration were smaller, although the N95 remained number one. She noted that the study results probably underestimate the true filtration efficacy of N95 masks, since they form a tighter seal with the face in clinical practice than with the other two mask types.

Mask self-protection “is easily applicable in your own practice, and it meets NIOSH-recommended [National Institute for Occupational Safety and Health] standards for safety in the workplace,” the dermatologist noted.

In the next phase of her research, she plans to evaluate the optimal technology and techniques of smoke evacuation in the surgical suite. That’s an attractive method because it protects everyone in the room, not just the surgeon. And while the practitioner survey indicates this technology isn’t widely used by dermatologic surgeons on a routine basis at present, that could change, particularly in the current era in which patient-reported outcomes and satisfaction surveys have taken on added weight.

 

 


“Patients prefer not smelling their own tissue burning,” Dr. de Golian said.

She reported no financial conflicts regarding her study, which was conducted free of commercial support.
 
Publications
Publications
Topics
Article Type
Click for Credit Status
Ready
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: Electrocautery smoke is bad news, and wearing an N95 mask affords protection.

Major finding: The N95 mask was significantly more effective than basic procedural or laser masks at filtering particulate matter less than 1 mcm in size contained in electrocautery smoke.

Study details: This study utilized highly sensitive airborne particle counting devices to assess the relative protective filtration afforded by three types of masks.

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

Disqus Comments
Default
Use ProPublica

Checkpoint inhibitors get to patients quickly

Article Type
Changed
Mon, 01/14/2019 - 10:23

 

Checkpoint inhibitors got to melanoma, non–small cell lung cancer, and renal cell carcinoma patients quickly in clinical practice after Food and Drug Administration approval – usually within 4 months – but the patients treated in clinical settings tended to be older than those treated in trials, which has caused concern about whether real-world efficacy will prove to be the same, according to a study in JAMA Oncology.

“Such rapid adoption stands in contrast to older estimates that suggest it takes years or even decades for new treatments to be adopted,” wrote lead author Cary Gross, MD, professor of medicine at Yale University, New Haven, and his coauthors. “We found significant differences in age between patients treated in practice and those treated in trials, which highlights the need to clarify the risks and benefits of checkpoint inhibitors in general populations of patients.”

Researchers drew data on nivolumab and pembrolizumab use from the Flatiron Health longitudinal EHR database, which included 233 academic and community oncology practices. In each of the three disease cohorts, adoption was seen within 4 months for at least 60% of patients. Uptake was quickest for the melanoma patients, 76% of whom received a checkpoint inhibitor within 4 months, investigators wrote. Factors for the fast adoption could include high disease severity, a preference for novelty, perceived gains over existing treatments, and promotional activities, such as media reports and advertising directly to consumers, they wrote.

More patients in real-world practice were aged 65 years or older, ranging from as little as 61% at the lowest end of the range at one center to as much as 64% at the highest end at another. In the clinical trials, the percentage of patients aged 65 years or older ranged from 32% in one trial to 41% in another. Researchers wrote that these higher real-world rates are concerning because there are still questions regarding whether differences in immune responses will cause differences in efficacy between older and younger patients, as well as safety considerations among different age groups.

“Although data suggest that outcomes are similar between older and younger patients receiving anti–PD-1 agents for melanoma, there is little evidence to guide anti–PD-1 treatment of older patients with NSCLC [non–small cell lung cancer],” Dr. Gross and his coinvestigators wrote.

Investigators wrote that the findings are cause for caution.

“As FDA officials develop more flexible standards for approval, which the 21st Century Cures Act requires them to do, it is possible that many patients will receive drugs before much is known about clinical outcomes,” Dr. Gross said. “Further integrations of real-world evidence might allow the FDA to better assess the drugs that they approve on the basis of nonrepresentative trial participants.”

SOURCE: Gross C et al. JAMA Oncol. 2018 May 10. doi: 10.1001/jamaoncol.2018.0798.






 

Publications
Topics
Sections

 

Checkpoint inhibitors got to melanoma, non–small cell lung cancer, and renal cell carcinoma patients quickly in clinical practice after Food and Drug Administration approval – usually within 4 months – but the patients treated in clinical settings tended to be older than those treated in trials, which has caused concern about whether real-world efficacy will prove to be the same, according to a study in JAMA Oncology.

“Such rapid adoption stands in contrast to older estimates that suggest it takes years or even decades for new treatments to be adopted,” wrote lead author Cary Gross, MD, professor of medicine at Yale University, New Haven, and his coauthors. “We found significant differences in age between patients treated in practice and those treated in trials, which highlights the need to clarify the risks and benefits of checkpoint inhibitors in general populations of patients.”

Researchers drew data on nivolumab and pembrolizumab use from the Flatiron Health longitudinal EHR database, which included 233 academic and community oncology practices. In each of the three disease cohorts, adoption was seen within 4 months for at least 60% of patients. Uptake was quickest for the melanoma patients, 76% of whom received a checkpoint inhibitor within 4 months, investigators wrote. Factors for the fast adoption could include high disease severity, a preference for novelty, perceived gains over existing treatments, and promotional activities, such as media reports and advertising directly to consumers, they wrote.

More patients in real-world practice were aged 65 years or older, ranging from as little as 61% at the lowest end of the range at one center to as much as 64% at the highest end at another. In the clinical trials, the percentage of patients aged 65 years or older ranged from 32% in one trial to 41% in another. Researchers wrote that these higher real-world rates are concerning because there are still questions regarding whether differences in immune responses will cause differences in efficacy between older and younger patients, as well as safety considerations among different age groups.

“Although data suggest that outcomes are similar between older and younger patients receiving anti–PD-1 agents for melanoma, there is little evidence to guide anti–PD-1 treatment of older patients with NSCLC [non–small cell lung cancer],” Dr. Gross and his coinvestigators wrote.

Investigators wrote that the findings are cause for caution.

“As FDA officials develop more flexible standards for approval, which the 21st Century Cures Act requires them to do, it is possible that many patients will receive drugs before much is known about clinical outcomes,” Dr. Gross said. “Further integrations of real-world evidence might allow the FDA to better assess the drugs that they approve on the basis of nonrepresentative trial participants.”

SOURCE: Gross C et al. JAMA Oncol. 2018 May 10. doi: 10.1001/jamaoncol.2018.0798.






 

 

Checkpoint inhibitors got to melanoma, non–small cell lung cancer, and renal cell carcinoma patients quickly in clinical practice after Food and Drug Administration approval – usually within 4 months – but the patients treated in clinical settings tended to be older than those treated in trials, which has caused concern about whether real-world efficacy will prove to be the same, according to a study in JAMA Oncology.

“Such rapid adoption stands in contrast to older estimates that suggest it takes years or even decades for new treatments to be adopted,” wrote lead author Cary Gross, MD, professor of medicine at Yale University, New Haven, and his coauthors. “We found significant differences in age between patients treated in practice and those treated in trials, which highlights the need to clarify the risks and benefits of checkpoint inhibitors in general populations of patients.”

Researchers drew data on nivolumab and pembrolizumab use from the Flatiron Health longitudinal EHR database, which included 233 academic and community oncology practices. In each of the three disease cohorts, adoption was seen within 4 months for at least 60% of patients. Uptake was quickest for the melanoma patients, 76% of whom received a checkpoint inhibitor within 4 months, investigators wrote. Factors for the fast adoption could include high disease severity, a preference for novelty, perceived gains over existing treatments, and promotional activities, such as media reports and advertising directly to consumers, they wrote.

More patients in real-world practice were aged 65 years or older, ranging from as little as 61% at the lowest end of the range at one center to as much as 64% at the highest end at another. In the clinical trials, the percentage of patients aged 65 years or older ranged from 32% in one trial to 41% in another. Researchers wrote that these higher real-world rates are concerning because there are still questions regarding whether differences in immune responses will cause differences in efficacy between older and younger patients, as well as safety considerations among different age groups.

“Although data suggest that outcomes are similar between older and younger patients receiving anti–PD-1 agents for melanoma, there is little evidence to guide anti–PD-1 treatment of older patients with NSCLC [non–small cell lung cancer],” Dr. Gross and his coinvestigators wrote.

Investigators wrote that the findings are cause for caution.

“As FDA officials develop more flexible standards for approval, which the 21st Century Cures Act requires them to do, it is possible that many patients will receive drugs before much is known about clinical outcomes,” Dr. Gross said. “Further integrations of real-world evidence might allow the FDA to better assess the drugs that they approve on the basis of nonrepresentative trial participants.”

SOURCE: Gross C et al. JAMA Oncol. 2018 May 10. doi: 10.1001/jamaoncol.2018.0798.






 

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM JAMA ONCOLOGY

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

 

Key clinical point: Fast adoption but older patient age is seen in clinical use of checkpoint inhibitors.

Major finding: About 60% of patients were aged 65 years and older in clinical settings, compared with about 30%-40% in trials.

Study details: A retrospective study of data from the Flatiron Health longitudinal EHR database, which includes 233 academic and community practices.

Disclosures: The study was sponsored by the National Institutes of Health. Investigators reported receiving financial support from Bristol-Myers Squibb, Eli Lilly, Johnson & Johnson, and other companies.

Source: Gross C et al. JAMA Oncology. 2018 May 10. doi: 10.1001/jamaoncol.2018.0798.

Disqus Comments
Default
Use ProPublica

Enhanced Melanoma Diagnosis With Multispectral Digital Skin Lesion Analysis

Article Type
Changed
Thu, 01/10/2019 - 13:50
Display Headline
Enhanced Melanoma Diagnosis With Multispectral Digital Skin Lesion Analysis

Early detection of melanoma, which is known to improve survival rates, remains a challenge for dermatologists. Suspicious pigmented lesions typically are evaluated via clinical examination and dermoscopy; however, new technologies are being developed to provide additional objective information for clinicians to incorporate into their biopsy decisions.

Multispectral digital skin lesion analysis (MSDSLA) uses 10 bands of visible and near-infrared light (430–950 nm) to image and analyze pigmented skin lesions (PSLs) down to 2.5 mm below the skin surface and measures the distribution of melanin using 75 unique algorithms to determine the degree of the morphologic disorder. Using a logical regression model previously validated on a set of 1632 PSLs, the probability of melanoma and probability of being a melanoma/PSL of high-risk malignant potential are then provided to the clinician.1

In this study, we analyzed aggregate data from 7 prior studies2-8 to better determine how MSDSLA impacts the biopsy decisions of dermatologists and nondermatologists following clinical examination and dermoscopic evaluation of PSLs.

Methods

A total of 855 practitioners (657 dermatologists, 126 dermatology residents, 72 nondermatologists [ie, primary care physicians, physician assistants, nurse practitioners]) in 7 prior reader studies (Table)2-8 were shown a total of 62 clinical (distant and close-up) and dermoscopic images of PSLs (13 invasive melanomas, 10 melanomas in situ, 7 high-grade dysplastic nevi, 32 benign skin lesions including low-grade dysplastic nevi) previously analyzed by MSDSLA.2-8 For each lesion evaluated, the practitioners were first asked if they would biopsy based on their review of the clinical and dermoscopic images and were asked again when given the associated MSDSLA information. Data were aggregated across all participants for the individual lesions presented in each reader study. Biopsy decisions were compared overall after evaluation of clinical and dermoscopic findings and then after evaluation of MSDSLA findings. Statistical analyses were performed using t-test and χ2 analysis for proportions where appropriate.

Results

Overall sensitivity for the detection of melanoma or other high-grade PSLs improved from 70% on clinical and dermoscopic evaluation to 88% after MSDSLA information was provided (P<.0001), and specificity increased from 52% to 58% (P<.001). Diagnostic accuracy also improved from 59% on clinical evaluation to 69% after review of MSDSLA findings (P<.0001). The positive predictive value of biopsy decisions was 47% following clinical evaluation, which improved to 56% after evaluation of MSDSLA findings (P<.001), and the negative predictive value increased from 74% to 89% (P<.0001). The overall percentage of lesions selected for biopsy did not significantly change following MSDSLA data integration (57% vs 60%)(Figure). Given that similar numbers of lesions were biopsied with improved sensitivity and specificity, the integration of MSDSLA data into the biopsy decision led to an improved biopsy ratio (ratio of melanomas biopsied to total biopsies) and fewer unnecessary biopsies.

Standard statistical metrics evaluating the impact of multispectral digital skin lesion analysis on pigmented lesion diagnosis. All 5 of the standard metrics for diagnostic tests improved following the provision of multispectral digital skin lesion analysis data to the health care providers (N=855). Asterisk indicates statiscally significant improvement (P< .05).

Comment

Our broad analysis further supported the findings of prior studies that decisions to biopsy clinically suspicious PSLs are more sensitive, specific, and accurate when practitioners are provided MSDSLA information following clinical examination.2-8With no significant increase in the number of biopsies performed, the fact that all 5 of the standard diagnostic evaluation metrics (sensitivity, specificity, diagnostic accuracy, positive predictive value, negative predictive value) were improved after MSDSLA information was provided additionally supported this conclusion.

Given the evolution in health care economics, it is clear that greater emphasis will continue to be placed on superior, evidence-based, effective care. The reported diagnostic sensitivities and specificities of clinical evaluation and dermoscopy for melanoma detection vary widely throughout the literature, with sensitivities ranging from 58% to over 90% and specificities ranging from 77% to 99%.9-11Diagnostic performance generally has been found to be higher among dermatologists than nondermatologists and is highest in specialized pigmented lesion clinics.12

Our study had several limitations. For this analysis to be more representative of lesion biopsy selection in the clinical setting, biopsy sensitivity (correctly identifying lesions appropriate for biopsy) vs melanoma sensitivity (identifying a lesion as melanoma) was used.13 The overall sensitivity found was within the range of prior studies,2-8 but this approach may have potentially led to a lower specificity due to an increased number of lesions biopsied. Additionally, the melanomas selected for these studies were early (malignant melanoma in situ or mean thickness of invasive malignant melanoma of 0.3 mm), and the nonmelanomas (including low-grade dysplastic nevi) were not necessarily diagnostically straightforward. This may have led to the clinical and dermoscopic sensitivity and specificity noted being lower than in some prior studies.9-11

The risk of missing a melanoma with MSDSLA devices has led manufacturers to strive for a high sensitivity for their devices, leading to lower specificity as a consequence. For this reason and other ambiguous practical considerations (eg, device and patient costs, difficulty with insurance reimbursement), the adoption of this technology into routine clinical practice has remained relatively static; however, using enhanced diagnostic technologies such as MSDSLA may help with more accurate identification of high-risk PSLs, thereby leading to earlier detection and overall less expensive, more cost-effective treatment of melanoma.

References
  1. Monheit G, Cognetta AB, Ferris L, et al. The performance of MelaFind: a prospective multicenter study. Arch Dermatol. 2011;147:188-194.
  2. Rigel DS, Roy M, Yoo J, et al. Impact of guidance from a computer-aided multispectral digital skin lesion analysis device on decision to biopsy lesions clinically suggestive of melanoma. Arch Dermatol. 2012;148:541-543.
  3. Yoo J, Rigel DS, Roy M, et al. Impact of guidance from a multispectral digital skin lesion analysis device on dermatology residents decisions to biopsy lesions clinically suggestive of melanoma. J Am Acad Dermatol. 2013;68:AB152.
  4. Winkelmann RR, Yoo J, Tucker N, et al. Impact of guidance provided by a multispectral digital skin lesion analysis device following dermoscopy on decisions to biopsy atypical melanocytic lesions. J Clin Aesthet Dermatol. 2015;8:21-24.
  5. Winkelmann RR, Hauschild A, Tucker N, et al. The impact of multispectral digital skin lesion analysis on German dermatologist decisions to biopsy atypical pigmented lesions with clinical characteristics of melanoma. J Clin Aesthet Dermatol. 2015;8:27-29.
  6. Winkelmann RR, Tucker N, White R, et al. Pigmented skin lesion biopsies after computer-aided multispectral digital skin lesion analysis. J Am Osteopath Assoc. 2015;115:666-669.
  7. Winkelmann RR, Farberg AS, Tucker N, et al. Enhancement of international dermatologists’ pigmented skin lesion biopsy decisions following dermoscopy with subsequent integration of multispectral digital skin lesion analysis [published online July 1, 2016]. J Clin Aesthet Dermatol. 2016;9:53-55.
  8. Farberg AS, Winkelmann RR, Tucker N, et al. The impact of quantitative data provided by a multi-spectral digital skin lesion analysis device on dermatologists’ decisions to biopsy pigmented lesions [published online September 1, 2017]. J Clin Aesthet Dermatol. 2017;10:24-26.
  9. Wolf IH, Smolle J, Soyer HP, et al. Sensitivity in the clinical diagnosis of malignant melanoma. Melanoma Res. 1998;8:425-429.
  10. Kittler H, Pehamberger H, Wolff K, et al. Diagnostic accuracy of dermoscopy. Lancet Oncol. 2002;3:159-165.
  11. Ascierto PA, Palmieri G, Celentano E, et al. Sensitivity and specificity of epiluminescence microscopy: evaluation on a sample of 2731 excised cutaneous pigmented lesions: the Melanoma Cooperative Study. Br J Dermatol. 2000;142:893-898.
  12. Carli P, Nardini P, Crocetti E, et al. Frequency and characteristics of melanomas missed at a pigmented lesion clinic: a registry-based study. Melanoma Res. 2004;14:403-407.
  13. Friedman RJ, Gutkowicz-Krusin D, Farber MJ, et al. The diagnostic performance of expert dermoscopists vs a computer-vision system on small-diameter melanomas. Arch Dermatol. 2008;144:476-482.
Article PDF
Author and Disclosure Information

Dr. Farberg is from the Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York. Dr. Glazer is from the Division of Dermatology, University of Arizona, Tucson. Dr. Winkelmann is from the Department of Dermatology, OhioHealth, Athens. Ms. Tucker is from STRATA Skin Sciences, Horsham, Pennsylvania. Mr. White is from IRIS Interactive Horizon Inc, Cody, Wyoming. Dr. Rigel is from the Department of Dermatology, New York University School of Medicine, New York.

Drs. Glazer and White report no conflict of interest. Drs. Farberg and Winkelman received research funding from STRATA Skin Sciences. Ms. Tucker is an employee of STRATA Skin Sciences. Dr. Rigel was a consultant for STRATA Skin Sciences.

Correspondence: Darrell S. Rigel, MD, MS, 35 E 35th St, #208, New York, NY, 10016 (dsrigel@prodigy.net).

Issue
Cutis - 101(5)
Publications
Topics
Page Number
338-340
Sections
Author and Disclosure Information

Dr. Farberg is from the Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York. Dr. Glazer is from the Division of Dermatology, University of Arizona, Tucson. Dr. Winkelmann is from the Department of Dermatology, OhioHealth, Athens. Ms. Tucker is from STRATA Skin Sciences, Horsham, Pennsylvania. Mr. White is from IRIS Interactive Horizon Inc, Cody, Wyoming. Dr. Rigel is from the Department of Dermatology, New York University School of Medicine, New York.

Drs. Glazer and White report no conflict of interest. Drs. Farberg and Winkelman received research funding from STRATA Skin Sciences. Ms. Tucker is an employee of STRATA Skin Sciences. Dr. Rigel was a consultant for STRATA Skin Sciences.

Correspondence: Darrell S. Rigel, MD, MS, 35 E 35th St, #208, New York, NY, 10016 (dsrigel@prodigy.net).

Author and Disclosure Information

Dr. Farberg is from the Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York. Dr. Glazer is from the Division of Dermatology, University of Arizona, Tucson. Dr. Winkelmann is from the Department of Dermatology, OhioHealth, Athens. Ms. Tucker is from STRATA Skin Sciences, Horsham, Pennsylvania. Mr. White is from IRIS Interactive Horizon Inc, Cody, Wyoming. Dr. Rigel is from the Department of Dermatology, New York University School of Medicine, New York.

Drs. Glazer and White report no conflict of interest. Drs. Farberg and Winkelman received research funding from STRATA Skin Sciences. Ms. Tucker is an employee of STRATA Skin Sciences. Dr. Rigel was a consultant for STRATA Skin Sciences.

Correspondence: Darrell S. Rigel, MD, MS, 35 E 35th St, #208, New York, NY, 10016 (dsrigel@prodigy.net).

Article PDF
Article PDF

Early detection of melanoma, which is known to improve survival rates, remains a challenge for dermatologists. Suspicious pigmented lesions typically are evaluated via clinical examination and dermoscopy; however, new technologies are being developed to provide additional objective information for clinicians to incorporate into their biopsy decisions.

Multispectral digital skin lesion analysis (MSDSLA) uses 10 bands of visible and near-infrared light (430–950 nm) to image and analyze pigmented skin lesions (PSLs) down to 2.5 mm below the skin surface and measures the distribution of melanin using 75 unique algorithms to determine the degree of the morphologic disorder. Using a logical regression model previously validated on a set of 1632 PSLs, the probability of melanoma and probability of being a melanoma/PSL of high-risk malignant potential are then provided to the clinician.1

In this study, we analyzed aggregate data from 7 prior studies2-8 to better determine how MSDSLA impacts the biopsy decisions of dermatologists and nondermatologists following clinical examination and dermoscopic evaluation of PSLs.

Methods

A total of 855 practitioners (657 dermatologists, 126 dermatology residents, 72 nondermatologists [ie, primary care physicians, physician assistants, nurse practitioners]) in 7 prior reader studies (Table)2-8 were shown a total of 62 clinical (distant and close-up) and dermoscopic images of PSLs (13 invasive melanomas, 10 melanomas in situ, 7 high-grade dysplastic nevi, 32 benign skin lesions including low-grade dysplastic nevi) previously analyzed by MSDSLA.2-8 For each lesion evaluated, the practitioners were first asked if they would biopsy based on their review of the clinical and dermoscopic images and were asked again when given the associated MSDSLA information. Data were aggregated across all participants for the individual lesions presented in each reader study. Biopsy decisions were compared overall after evaluation of clinical and dermoscopic findings and then after evaluation of MSDSLA findings. Statistical analyses were performed using t-test and χ2 analysis for proportions where appropriate.

Results

Overall sensitivity for the detection of melanoma or other high-grade PSLs improved from 70% on clinical and dermoscopic evaluation to 88% after MSDSLA information was provided (P<.0001), and specificity increased from 52% to 58% (P<.001). Diagnostic accuracy also improved from 59% on clinical evaluation to 69% after review of MSDSLA findings (P<.0001). The positive predictive value of biopsy decisions was 47% following clinical evaluation, which improved to 56% after evaluation of MSDSLA findings (P<.001), and the negative predictive value increased from 74% to 89% (P<.0001). The overall percentage of lesions selected for biopsy did not significantly change following MSDSLA data integration (57% vs 60%)(Figure). Given that similar numbers of lesions were biopsied with improved sensitivity and specificity, the integration of MSDSLA data into the biopsy decision led to an improved biopsy ratio (ratio of melanomas biopsied to total biopsies) and fewer unnecessary biopsies.

Standard statistical metrics evaluating the impact of multispectral digital skin lesion analysis on pigmented lesion diagnosis. All 5 of the standard metrics for diagnostic tests improved following the provision of multispectral digital skin lesion analysis data to the health care providers (N=855). Asterisk indicates statiscally significant improvement (P< .05).

Comment

Our broad analysis further supported the findings of prior studies that decisions to biopsy clinically suspicious PSLs are more sensitive, specific, and accurate when practitioners are provided MSDSLA information following clinical examination.2-8With no significant increase in the number of biopsies performed, the fact that all 5 of the standard diagnostic evaluation metrics (sensitivity, specificity, diagnostic accuracy, positive predictive value, negative predictive value) were improved after MSDSLA information was provided additionally supported this conclusion.

Given the evolution in health care economics, it is clear that greater emphasis will continue to be placed on superior, evidence-based, effective care. The reported diagnostic sensitivities and specificities of clinical evaluation and dermoscopy for melanoma detection vary widely throughout the literature, with sensitivities ranging from 58% to over 90% and specificities ranging from 77% to 99%.9-11Diagnostic performance generally has been found to be higher among dermatologists than nondermatologists and is highest in specialized pigmented lesion clinics.12

Our study had several limitations. For this analysis to be more representative of lesion biopsy selection in the clinical setting, biopsy sensitivity (correctly identifying lesions appropriate for biopsy) vs melanoma sensitivity (identifying a lesion as melanoma) was used.13 The overall sensitivity found was within the range of prior studies,2-8 but this approach may have potentially led to a lower specificity due to an increased number of lesions biopsied. Additionally, the melanomas selected for these studies were early (malignant melanoma in situ or mean thickness of invasive malignant melanoma of 0.3 mm), and the nonmelanomas (including low-grade dysplastic nevi) were not necessarily diagnostically straightforward. This may have led to the clinical and dermoscopic sensitivity and specificity noted being lower than in some prior studies.9-11

The risk of missing a melanoma with MSDSLA devices has led manufacturers to strive for a high sensitivity for their devices, leading to lower specificity as a consequence. For this reason and other ambiguous practical considerations (eg, device and patient costs, difficulty with insurance reimbursement), the adoption of this technology into routine clinical practice has remained relatively static; however, using enhanced diagnostic technologies such as MSDSLA may help with more accurate identification of high-risk PSLs, thereby leading to earlier detection and overall less expensive, more cost-effective treatment of melanoma.

Early detection of melanoma, which is known to improve survival rates, remains a challenge for dermatologists. Suspicious pigmented lesions typically are evaluated via clinical examination and dermoscopy; however, new technologies are being developed to provide additional objective information for clinicians to incorporate into their biopsy decisions.

Multispectral digital skin lesion analysis (MSDSLA) uses 10 bands of visible and near-infrared light (430–950 nm) to image and analyze pigmented skin lesions (PSLs) down to 2.5 mm below the skin surface and measures the distribution of melanin using 75 unique algorithms to determine the degree of the morphologic disorder. Using a logical regression model previously validated on a set of 1632 PSLs, the probability of melanoma and probability of being a melanoma/PSL of high-risk malignant potential are then provided to the clinician.1

In this study, we analyzed aggregate data from 7 prior studies2-8 to better determine how MSDSLA impacts the biopsy decisions of dermatologists and nondermatologists following clinical examination and dermoscopic evaluation of PSLs.

Methods

A total of 855 practitioners (657 dermatologists, 126 dermatology residents, 72 nondermatologists [ie, primary care physicians, physician assistants, nurse practitioners]) in 7 prior reader studies (Table)2-8 were shown a total of 62 clinical (distant and close-up) and dermoscopic images of PSLs (13 invasive melanomas, 10 melanomas in situ, 7 high-grade dysplastic nevi, 32 benign skin lesions including low-grade dysplastic nevi) previously analyzed by MSDSLA.2-8 For each lesion evaluated, the practitioners were first asked if they would biopsy based on their review of the clinical and dermoscopic images and were asked again when given the associated MSDSLA information. Data were aggregated across all participants for the individual lesions presented in each reader study. Biopsy decisions were compared overall after evaluation of clinical and dermoscopic findings and then after evaluation of MSDSLA findings. Statistical analyses were performed using t-test and χ2 analysis for proportions where appropriate.

Results

Overall sensitivity for the detection of melanoma or other high-grade PSLs improved from 70% on clinical and dermoscopic evaluation to 88% after MSDSLA information was provided (P<.0001), and specificity increased from 52% to 58% (P<.001). Diagnostic accuracy also improved from 59% on clinical evaluation to 69% after review of MSDSLA findings (P<.0001). The positive predictive value of biopsy decisions was 47% following clinical evaluation, which improved to 56% after evaluation of MSDSLA findings (P<.001), and the negative predictive value increased from 74% to 89% (P<.0001). The overall percentage of lesions selected for biopsy did not significantly change following MSDSLA data integration (57% vs 60%)(Figure). Given that similar numbers of lesions were biopsied with improved sensitivity and specificity, the integration of MSDSLA data into the biopsy decision led to an improved biopsy ratio (ratio of melanomas biopsied to total biopsies) and fewer unnecessary biopsies.

Standard statistical metrics evaluating the impact of multispectral digital skin lesion analysis on pigmented lesion diagnosis. All 5 of the standard metrics for diagnostic tests improved following the provision of multispectral digital skin lesion analysis data to the health care providers (N=855). Asterisk indicates statiscally significant improvement (P< .05).

Comment

Our broad analysis further supported the findings of prior studies that decisions to biopsy clinically suspicious PSLs are more sensitive, specific, and accurate when practitioners are provided MSDSLA information following clinical examination.2-8With no significant increase in the number of biopsies performed, the fact that all 5 of the standard diagnostic evaluation metrics (sensitivity, specificity, diagnostic accuracy, positive predictive value, negative predictive value) were improved after MSDSLA information was provided additionally supported this conclusion.

Given the evolution in health care economics, it is clear that greater emphasis will continue to be placed on superior, evidence-based, effective care. The reported diagnostic sensitivities and specificities of clinical evaluation and dermoscopy for melanoma detection vary widely throughout the literature, with sensitivities ranging from 58% to over 90% and specificities ranging from 77% to 99%.9-11Diagnostic performance generally has been found to be higher among dermatologists than nondermatologists and is highest in specialized pigmented lesion clinics.12

Our study had several limitations. For this analysis to be more representative of lesion biopsy selection in the clinical setting, biopsy sensitivity (correctly identifying lesions appropriate for biopsy) vs melanoma sensitivity (identifying a lesion as melanoma) was used.13 The overall sensitivity found was within the range of prior studies,2-8 but this approach may have potentially led to a lower specificity due to an increased number of lesions biopsied. Additionally, the melanomas selected for these studies were early (malignant melanoma in situ or mean thickness of invasive malignant melanoma of 0.3 mm), and the nonmelanomas (including low-grade dysplastic nevi) were not necessarily diagnostically straightforward. This may have led to the clinical and dermoscopic sensitivity and specificity noted being lower than in some prior studies.9-11

The risk of missing a melanoma with MSDSLA devices has led manufacturers to strive for a high sensitivity for their devices, leading to lower specificity as a consequence. For this reason and other ambiguous practical considerations (eg, device and patient costs, difficulty with insurance reimbursement), the adoption of this technology into routine clinical practice has remained relatively static; however, using enhanced diagnostic technologies such as MSDSLA may help with more accurate identification of high-risk PSLs, thereby leading to earlier detection and overall less expensive, more cost-effective treatment of melanoma.

References
  1. Monheit G, Cognetta AB, Ferris L, et al. The performance of MelaFind: a prospective multicenter study. Arch Dermatol. 2011;147:188-194.
  2. Rigel DS, Roy M, Yoo J, et al. Impact of guidance from a computer-aided multispectral digital skin lesion analysis device on decision to biopsy lesions clinically suggestive of melanoma. Arch Dermatol. 2012;148:541-543.
  3. Yoo J, Rigel DS, Roy M, et al. Impact of guidance from a multispectral digital skin lesion analysis device on dermatology residents decisions to biopsy lesions clinically suggestive of melanoma. J Am Acad Dermatol. 2013;68:AB152.
  4. Winkelmann RR, Yoo J, Tucker N, et al. Impact of guidance provided by a multispectral digital skin lesion analysis device following dermoscopy on decisions to biopsy atypical melanocytic lesions. J Clin Aesthet Dermatol. 2015;8:21-24.
  5. Winkelmann RR, Hauschild A, Tucker N, et al. The impact of multispectral digital skin lesion analysis on German dermatologist decisions to biopsy atypical pigmented lesions with clinical characteristics of melanoma. J Clin Aesthet Dermatol. 2015;8:27-29.
  6. Winkelmann RR, Tucker N, White R, et al. Pigmented skin lesion biopsies after computer-aided multispectral digital skin lesion analysis. J Am Osteopath Assoc. 2015;115:666-669.
  7. Winkelmann RR, Farberg AS, Tucker N, et al. Enhancement of international dermatologists’ pigmented skin lesion biopsy decisions following dermoscopy with subsequent integration of multispectral digital skin lesion analysis [published online July 1, 2016]. J Clin Aesthet Dermatol. 2016;9:53-55.
  8. Farberg AS, Winkelmann RR, Tucker N, et al. The impact of quantitative data provided by a multi-spectral digital skin lesion analysis device on dermatologists’ decisions to biopsy pigmented lesions [published online September 1, 2017]. J Clin Aesthet Dermatol. 2017;10:24-26.
  9. Wolf IH, Smolle J, Soyer HP, et al. Sensitivity in the clinical diagnosis of malignant melanoma. Melanoma Res. 1998;8:425-429.
  10. Kittler H, Pehamberger H, Wolff K, et al. Diagnostic accuracy of dermoscopy. Lancet Oncol. 2002;3:159-165.
  11. Ascierto PA, Palmieri G, Celentano E, et al. Sensitivity and specificity of epiluminescence microscopy: evaluation on a sample of 2731 excised cutaneous pigmented lesions: the Melanoma Cooperative Study. Br J Dermatol. 2000;142:893-898.
  12. Carli P, Nardini P, Crocetti E, et al. Frequency and characteristics of melanomas missed at a pigmented lesion clinic: a registry-based study. Melanoma Res. 2004;14:403-407.
  13. Friedman RJ, Gutkowicz-Krusin D, Farber MJ, et al. The diagnostic performance of expert dermoscopists vs a computer-vision system on small-diameter melanomas. Arch Dermatol. 2008;144:476-482.
References
  1. Monheit G, Cognetta AB, Ferris L, et al. The performance of MelaFind: a prospective multicenter study. Arch Dermatol. 2011;147:188-194.
  2. Rigel DS, Roy M, Yoo J, et al. Impact of guidance from a computer-aided multispectral digital skin lesion analysis device on decision to biopsy lesions clinically suggestive of melanoma. Arch Dermatol. 2012;148:541-543.
  3. Yoo J, Rigel DS, Roy M, et al. Impact of guidance from a multispectral digital skin lesion analysis device on dermatology residents decisions to biopsy lesions clinically suggestive of melanoma. J Am Acad Dermatol. 2013;68:AB152.
  4. Winkelmann RR, Yoo J, Tucker N, et al. Impact of guidance provided by a multispectral digital skin lesion analysis device following dermoscopy on decisions to biopsy atypical melanocytic lesions. J Clin Aesthet Dermatol. 2015;8:21-24.
  5. Winkelmann RR, Hauschild A, Tucker N, et al. The impact of multispectral digital skin lesion analysis on German dermatologist decisions to biopsy atypical pigmented lesions with clinical characteristics of melanoma. J Clin Aesthet Dermatol. 2015;8:27-29.
  6. Winkelmann RR, Tucker N, White R, et al. Pigmented skin lesion biopsies after computer-aided multispectral digital skin lesion analysis. J Am Osteopath Assoc. 2015;115:666-669.
  7. Winkelmann RR, Farberg AS, Tucker N, et al. Enhancement of international dermatologists’ pigmented skin lesion biopsy decisions following dermoscopy with subsequent integration of multispectral digital skin lesion analysis [published online July 1, 2016]. J Clin Aesthet Dermatol. 2016;9:53-55.
  8. Farberg AS, Winkelmann RR, Tucker N, et al. The impact of quantitative data provided by a multi-spectral digital skin lesion analysis device on dermatologists’ decisions to biopsy pigmented lesions [published online September 1, 2017]. J Clin Aesthet Dermatol. 2017;10:24-26.
  9. Wolf IH, Smolle J, Soyer HP, et al. Sensitivity in the clinical diagnosis of malignant melanoma. Melanoma Res. 1998;8:425-429.
  10. Kittler H, Pehamberger H, Wolff K, et al. Diagnostic accuracy of dermoscopy. Lancet Oncol. 2002;3:159-165.
  11. Ascierto PA, Palmieri G, Celentano E, et al. Sensitivity and specificity of epiluminescence microscopy: evaluation on a sample of 2731 excised cutaneous pigmented lesions: the Melanoma Cooperative Study. Br J Dermatol. 2000;142:893-898.
  12. Carli P, Nardini P, Crocetti E, et al. Frequency and characteristics of melanomas missed at a pigmented lesion clinic: a registry-based study. Melanoma Res. 2004;14:403-407.
  13. Friedman RJ, Gutkowicz-Krusin D, Farber MJ, et al. The diagnostic performance of expert dermoscopists vs a computer-vision system on small-diameter melanomas. Arch Dermatol. 2008;144:476-482.
Issue
Cutis - 101(5)
Issue
Cutis - 101(5)
Page Number
338-340
Page Number
338-340
Publications
Publications
Topics
Article Type
Display Headline
Enhanced Melanoma Diagnosis With Multispectral Digital Skin Lesion Analysis
Display Headline
Enhanced Melanoma Diagnosis With Multispectral Digital Skin Lesion Analysis
Sections
Inside the Article

Practice Points

  • Multispectral digital skin lesion analysis (MSDSLA) can be a valuable tool in the evaluation of pigmented skin lesions (PSLs).
  • MSDSLA may help to better identify high-risk PSLs and improve cost of care.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
PubMed ID
29894523
Disqus Comments
Default
Use ProPublica
Article PDF Media