Purpose: The emergence of skin tumors in patients treated with sorafenib or with more recent BRAF inhibitors is an intriguing and potentially serious event. We carried out a clinical, pathologic, and molecular study of skin lesions occurring in patients receiving sorafenib.

Experimental Design: Thirty-one skin lesions from patients receiving sorafenib were characterized clinically and pathologically. DNA extracted from the lesions was screened for mutation hot spots of HRAS, NRAS, KiRAS, TP53, EGFR, BRAF, AKT1, PI3KCA, TGFBR1, and PTEN. Biological effect of sorafenib was studied in vivo in normal skin specimen and in vitro on cultured keratinocytes.

Results: We observed a continuous spectrum of lesions: from benign to more inflammatory and proliferative lesions, all seemingly initiated in the hair follicles. Eight oncogenic HRAS, TGFBR1, and TP53 mutations were found in 2 benign lesions, 3 keratoacanthomas (KA) and 3 KA-like squamous cell carcinoma (SCC). Six of them correspond to the typical UV signature. Treatment with sorafenib led to an increased keratinocyte proliferation and a tendency toward increased mitogen-activated protein kinase (MAPK) pathway activation in normal skin.

Sorafenib induced BRAF–CRAF dimerization in cultured keratinocytes and activated CRAF with a dose-dependent effect on MAP-kinase pathway activation and on keratinocyte proliferation.

Conclusion: Sorafenib induces keratinocyte proliferation in vivo and a time- and dose-dependent activation of the MAP kinase pathway in vitro. It is associated with a spectrum of lesions ranging from benign follicular cystic lesions to KA-like SCC. Additional and potentially preexisting somatic genetic events, like UV-induced mutations, might influence the evolution of benign lesions to more proliferative and malignant tumors. Clin Cancer Res; 18(1); 263–72. ©2011 AACR.

This article is featured in Highlights of This Issue, p. 1

Translational Relevance

Skin tumors appearing during the course of RAF inhibitors–based therapy is an intriguing and potentially serious adverse effect of these new drugs. We carried out a detailed clinical, pathologic, and molecular study of skin tumors in patients treated with sorafenib. In parallel, we studied the effect of sorafenib on normal keratinocytes in vivo in normal skin samples and in vitro. This translational study showed that the RAF inhibitor sorafenib induces a time- and dose-dependent paradoxic activation of the MAP kinase pathway in keratinocytes, leading to cell proliferation and associated with the emergence of benign (follicular cystic lesions), borderline (keratoacanthomas), and malignant (squamous cell carcinomas) tumors. Additional somatic events like oncogene mutations, possibly induced by UV rays exposure, might influence the evolution of benign lesions to malignant tumors. Understanding the mechanisms underlying this phenomenon is a first and mandatory step toward treatment and prevention of this worrisome side effect of our new targeted therapies.

BRAF is the most frequently mutated protein kinase in human cancer (1) and is the target of several anticancer drugs. The potency and the specificity of BRAF inhibitors available on the market or under clinical development are variable. Sorafenib (Nexavar, Bayer/Onyx) is a pan-RAF inhibitor which also blocks VEGF receptors (VEGFR)-2, -3, platelet-derived growth factor receptor-β, fms-like tyrosine kinase 3 (FLT3), and KIT. Conversely, vemurafenib (Plexxikon/Roche) or GSK2118436 (GlaxoSmithKline) are highly selective and very potent BRAF inhibitors. These latter drugs have a low IC50 for mutated oncogenic BRAF and are less active on wild-type BRAF (wtBRAF) and other RAF kinases. The clinical activities of these inhibitors also differ from one another. Selective BRAF inhibitors like vemurafenib and GSK2118436 are efficient against tumors harboring BRAF mutations and dependent on the RAF/MEK/ERK (mitogen-activated protein (MAP)/extracellular signal-regulated (ERK; MEK) kinase) pathway. Melanoma with V600E BRAF mutation is a case in point (2). Sorafenib activity is not linked to the presence of a BRAF mutation in the tumor and is more likely due to its antiangiogenic effect. Sorafenib is efficient and has been approved for the treatment of metastatic renal carcinoma (3, 4) and advanced hepatocellular carcinoma (4) independently of the mutational status of BRAF.

In spite of their variability in terms of BRAF selectivity and clinical activity, all 3 RAF inhibitors are associated with one and the same intriguing cutaneous side effect which is the emergence of borderline squamous cell neoplasms. Indeed, we and others have reported single or multiple cutaneous keratoacanthomas (KA) and squamous cell carcinomas (SCC) during treatment with sorafenib (5–9). These paradoxic keratinocyte proliferations arise in 6% to 7% of patients (5,10). The induction of KA and SCC is even more frequent with vemurafenib and GSK2118436 and has been described in 15% to 25% of the patients (2, 10).

We undertook a translational research in the aim of understanding the mechanisms of these skin cancer inductions. We first conducted a detailed clinical, pathologic, and molecular study of the skin neoplasms arising in patients treated with sorafenib. We then looked at the biological effects of sorafenib in vivo on normal skin samples from patients treated with sorafenib or a placebo. In parallel, we analyzed the effect of sorafenib in vitro on cultured keratinocytes. We found that sorafenib exposure was associated with increased mitogen-activated protein kinase (MAPK) pathway activation and keratinocyte proliferation ex vivo and in vivo. Sorafenib induced dose-dependent BRAF/CRAF dimerization associated with strong activation of CRAF in cultured keratinocytes. Finally, oncogenic mutations in HRAS, TGFBR1, and TP53 were detected in several benign or malignant skin tumors. Altogether, our results suggest that sorafenib induces a continuous spectrum of keratinocyte changes and proliferation via the paradoxic activation of the MAPK pathway in keratinocytes associated with BRAF/CRAF heterodimerization and subsequent CRAF activation. Additional events such as a HRAS mutation or EGFR activation giving rise to MAPK pathway coactivation might be required for full transformation of keratinocytes.

Patients and skin samples

Thirty-one skin lesions diagnosed as follicular cystic skin lesions, perforating folliculitis (PF), KA, or SCC originating from 17 patients treated with sorafenib from 2005 to 2010 were studied.

For each patient, the tumor type, duration of sorafenib therapy before the emergence of the skin tumor, and the clinical description of the skin lesions are reported. Four-micrometer sections from formalin-fixed, paraffin-embedded biopsy samples of skin lesions were stained with hematoxylin and eosin and independently read by 2 pathologists.

Normal skin samples from patients included in the TARGET study, evaluating sorafenib versus a placebo in patients with renal cell cancer were also studied (3). Normal skin biopsy samples were collected from the forearms of patients who received sorafenib or a placebo for 6 weeks.

Each skin sample was cut into 4-μm sections, formalin-fixed, paraffin-embedded, and stained with hematoxylin and eosin. A section from each case was analyzed immunochemically for Ki-67 (monoclonal MIB-1; Dako) and phospho-ERK protein (monoclonal 20G11; Cell Signaling Technology). Slides were deparaffinized and treated with an alcohol gradient to absolute ethanol. Endogenous peroxidase activity was blocked with the Peroxidase-Blocking Solution (Dako; S2001). Antigen retrieval was achieved with Target Retrieval Solution (Dako). Primary antibody was applied at 1:100 dilution and incubated at 4°C for 1 hour. Anti-mouse immunoglobulin G with the Dako EnVision System (horseradish peroxidase) was employed according to the manufacturer's instructions.

DNA isolation and mutation detection on biopsy specimens of skin lesions

Genomic DNA was extracted from 10- to 20-μm thick paraffin-embedded unstained slides of each skin lesion with the DNeasy Blood and Tissue Kit (QIAGEN) according to the manufacturer's instructions. We analyzed full coding sequences of 70 exons including oncogenic mutational hot spots corresponding to AKT1 (NM_005163.2) exon 4, BRAF (NM_004333.4) exons 11 and 15, EGFR (NM_005228.3) exons 18 to 21, HRAS (NM_005343.2) exons 2 and 3, KRAS (NM_033360.2) exons 2 and 3, NRAS (NM_002524.3) exon 2 and 3, PIK3CA (NM_006218.2) exons 10 and 21, PTEN (NM_000314.4) exons 1 to 9, TP53 (NM_000546.4) exons 2 and 11, and TGFBR1 (NM_004612.2) exons 2, 4, 6, and 7.

Sequences were analyzed by Sanger direct sequencing conducted after PCR amplification of target exons. Sequencing reactions were carried out by the BigDye Terminator Cycle Sequencing Kit software according to the manufacturer's recommendations (Applied Biosystems). The primer sequences are available upon request. Sequencing reactions were analyzed on a 48-capillary 3730 DNA Analyzer. Sequence reading and alignment were carried out with SeqScape software (Applied Biosystems). All detected mutations were confirmed following at least 1 independent PCR reaction.

In vitro experiment with HaCat keratinocytes

Cell culture.

HaCat keratinocytes were cultured in Dulbecco's Modified Eagle's Medium (Invitrogen) containing 10% fetal calf serum (Perbio) and antibiotics (penicillin/streptomycin; Invitrogen). Cellular proliferation was evaluated by the CellTiter Assay (Promega). Sorafenib and PD153035 used for cell treatment was purchased from Alexis Biochemicals and dissolved in dimethyl sulfoxide (DMSO).

Protein expression.

Cells were lysed in 50 mmol/L Tris (pH 7.5), 150 mmol/L NaCl, 0,5% NP40, 5 mmol/L NaF, 2 mmol/L Na3VO4, 10 μg/mL leupeptin, and 10 μg/mL aprotinin. For immunoprecipitation, samples were incubated with 1 μg BRAF (clones F-7 or H145; Santa Cruz) or CRAF (clone C12; Santa Cruz or clone 53/c-Raf-1; BD Transduction Laboratories) antibodies for 1 hour and immunoprecipitated by protein G-Sepharose (GE Healthcare) for 1 hour at 4°C. The immune complexes were washed 3 times with lysis buffer, proteins were separated by SDS-PAGE, and Western blot analysis was conducted according to standard protocols with the following antibodies: BRAF (F7 or H145); CRAF (C12 or 53/c-Raf-1); phospho-ERK (MAPK-YT; Sigma) and ERK (Upstate/Millipore); MCL1 (Abcam), EGFR, and phospho-EGFR (Cell Signaling); and Actin (Abcam).

Kinase assay.

CRAF was immunoprecipitated from cellular extract and its activity toward MEK measured in vitro. Briefly, immunoprecipitated CRAF was incubated with 5 μg of purified GST-MEK in 30 mmol/L Tris (pH 7.5), 100 μmol/L EDTA, 10 mmol/L MgCl2, 0.1% Triton X-100, 5 mmol/L NaF, 1 mmol/L ATP, 0.3% β-mercaptoethanol for 30 minutes at 30°C. The reaction was stopped by adding Laemmli buffer and analyzed by SDS-PAGE with the phospho-MEK (Cell Signaling Technology) and CRAF antibody. Phosphorylated MEK and CRAF were visualized and quantified by fluorescence on an Odyssey imaging system.

Proximity ligation assay.

HaCat cells were grown on glass cover slips, treated with DMSO or sorafenib, fixed with methanol, and permeabilized with acetone. The proximity ligation assay (PLA) was conducted according to the manufacturer's instructions (Olink) with B-Raf (F7) and C-Raf (C12) antibodies.

RNA interference.

HaCat cells were transfected with an siRNA control or targeting BRAF or CRAF as previously described (11).

Statistics.

Simple univariate descriptive statistics were used to compare the population, using the t test with a 5% significance level.

Clinical and pathologic characterization of skin lesions

The gender, age, cancer type, daily dose of sorafenib, time interval between the start of sorafenib therapy and the first skin symptom, the number and types of lesions as well as the skin sites involved are detailed in Table 1.

Table 1.

Patients and cutaneous lesions characteristics

PatientGender (M, male; F, female)AgeCancerDelay monthsLesionsSiteMutationCystic lesions
65 RCC 1 KA Nose TGFBR1-P83L Face 
49 RCC 1 KA Thigh WT Face 
44 RCC 1 KA Cheek TP53-G293R Face 
76 RCC 1 KA Forearm WT Axillary, genitals 
     Cystic folliculitis Axilla WT  
      Vulva NT  
     1 in situ SCC Cheek TP53-C277F  
70 RCC 1 KA Forearm NT Face 
74 HCC 1 KA Cheek WT Face 
     1 KA Cheek WT  
     1 KA Elbow HRAS-G13V  
69 RCC 1 KA-like SCC Palm HRAS- G13R Face, buttocks 
     Multiple KAs Dorsal hand WT  
      Neck WT  
      Torso WT  
     PF Buttocks WT  
51 MM 1 KA-like SCC Cheek WT Face 
79 RCC 1 KA-like SCC Back WT Face 
10 46 RCC 1 KA-like SCC Nose TGFBR1-G374R Face 
     Cystic folliculitis Nose WT  
11 57 RCC PF Buttocks WT Face, buttocks 
     Cystic folliculitis Face WT  
     1 KA Elbow WT  
12 60 RCC PF Buttocks TP53-R181H Face, buttocks 
13 41 MM PF Buttocks NT Face, buttocks 
14 62 RCC PF Legs WT Face, buttocks 
     PF Neck HRAS-G12S+G12D  
15 41 MM Multiple cystic folliculitis Buttocks WT Face, buttocks 
16  RCC Multiple cystic folliculitis Thigh and trunk NT Legs, trunk buttocks 
17 50 RCC Multiple cystic folliculitis Face WT Face, buttocks 
PatientGender (M, male; F, female)AgeCancerDelay monthsLesionsSiteMutationCystic lesions
65 RCC 1 KA Nose TGFBR1-P83L Face 
49 RCC 1 KA Thigh WT Face 
44 RCC 1 KA Cheek TP53-G293R Face 
76 RCC 1 KA Forearm WT Axillary, genitals 
     Cystic folliculitis Axilla WT  
      Vulva NT  
     1 in situ SCC Cheek TP53-C277F  
70 RCC 1 KA Forearm NT Face 
74 HCC 1 KA Cheek WT Face 
     1 KA Cheek WT  
     1 KA Elbow HRAS-G13V  
69 RCC 1 KA-like SCC Palm HRAS- G13R Face, buttocks 
     Multiple KAs Dorsal hand WT  
      Neck WT  
      Torso WT  
     PF Buttocks WT  
51 MM 1 KA-like SCC Cheek WT Face 
79 RCC 1 KA-like SCC Back WT Face 
10 46 RCC 1 KA-like SCC Nose TGFBR1-G374R Face 
     Cystic folliculitis Nose WT  
11 57 RCC PF Buttocks WT Face, buttocks 
     Cystic folliculitis Face WT  
     1 KA Elbow WT  
12 60 RCC PF Buttocks TP53-R181H Face, buttocks 
13 41 MM PF Buttocks NT Face, buttocks 
14 62 RCC PF Legs WT Face, buttocks 
     PF Neck HRAS-G12S+G12D  
15 41 MM Multiple cystic folliculitis Buttocks WT Face, buttocks 
16  RCC Multiple cystic folliculitis Thigh and trunk NT Legs, trunk buttocks 
17 50 RCC Multiple cystic folliculitis Face WT Face, buttocks 

▪ UV signature.

⊡ Non-UV induced.

Details of the mutated sequences:

TP53-R181H for mutation in exon 5 of TP53 (NM_000546.6) encoded c.542G>A; p.Arg181His.

TP53-C277F for mutation in exon 8 of TP53 (NM_000546.6) encoded c.830G>T; p.Cys277Phe.

TP53-G293R for mutation in exon 8 of TP53 (NM_000546.6) encoded c.877G>A; p.Gly293Arg.

HRAS-G13V for mutation in exon 2 of HRAS (NM_005343.2) encoded c.38G>T; p.Gly13Val.

HRAS-G13R for mutation in exon 2 of HRAS (NM_005343.2) encoded c.37G>C; p.Gly13Arg.

HRAS-G12S+G12D for double mutation in exon 2 of HRAS (NM_005343.2) encoded c.34G>A and c.35G>A; p.Gly12Ser and p.Gly12ASP.

TGFBR1-P83L for mutation in exon 2 of TGFBR1 (NM_004612.2) encoded c.248C>T; p.Pro83Leu.

TGFBR1-G374R for a mutation in exon 6 of TGFBR1 (NM_004612.2) encoded c.1120G>A; p.Gly374Arg.

Delay (month): Interval in months between the initiation of sorafenib and the emergence of the first skin lesion.

Abbreviations: WT, wild type; NT, not tested.

Various clinical aspects were observed but all patients developed small cystic follicular lesions that most frequently affected the face and also sometimes other parts of the skin such as the buttocks, axilla, or inguinal areas (Table 1). They occurred 3 to 5 months after the initiation of sorafenib. Cysts on the face were dominantly noninflammatory millimetric small and white lesions. Inflammatory cystic lesions exhibited erythematous hyperkeratotic papules, with a central, cone-shaped plug.

Lesions clinically suggestive of perforating folliculitis, meaning a more inflammatory infiltrate with a keratotic appearance occurred mostly on the buttocks and the upper part of the limbs (patients # 7, 11, 12, 13, and 14).

Patients 1 to 11 developed at least one lesion characteristic of KA or of KA-like SCC. Clinically, they were fast-growing firm, round nodules with a central crateriform keratotic zone. Three of these patients had multiple skin lesions (patients # 4, 6, and 7) occurring 1 to 9 months after the initiation of sorafenib.

A total of 22 lesions were excised and the pathologic examination with hematoxylin and eosin staining revealed a spectrum of lesions (Fig. 1): (i) noninflammatory cystic hair follicles, (ii) cystic folliculitis and perforating folliculitis with rupture of the hair follicle wall and perifollicular infiltrates, (iii) KA defined as a crateriform epidermal neoplasm limited by sharp beaks with no cytologic atypia nor invasion, (iv) crateriform KA-like SCC defined as crateriform epidermal neoplasms with irregular budding borders, cytologic atypia, and invasive nests (Fig. 1). Lymphatic metastatic embols were seen in one SCC (patient #8).

Figure 1.

Clinical and pathologic spectrum of skin lesions. Noninflammatory cystic follicular lesions (A and E), inflamed follicular cysts: folliculitis with onset of perforation (B and F), perforating folliculitis (C and G), and keratoacanthoma (D and H).

Figure 1.

Clinical and pathologic spectrum of skin lesions. Noninflammatory cystic follicular lesions (A and E), inflamed follicular cysts: folliculitis with onset of perforation (B and F), perforating folliculitis (C and G), and keratoacanthoma (D and H).

Close modal

Patient #7 developed the entire spectrum of the skin lesions that were observed: noninflammatory facial cysts, cystic folliculitis, perforating folliculitis, multiple KA, and SCC that emerged 3 to 5 months, respectively, after the initiation of sorafenib. Sorafenib was stopped due to cutaneous intolerance at the request of the patient after 24 months even though it was still effective against the progression of the renal cell carcinoma. All of her skin lesions, including skin tumors disappeared over a 4-month period following sorafenib interruption.

Oncogenic mutations

Eight oncogenic mutations were found in benign and malignant tumors. Three HRAS mutations on codon 12 or 13 were found in 3 lesions: 1 KA, 1 KA-like SCC, and 1 case of benign perforating folliculitis (Table 1). TP53 was also found to be mutated in 3 lesions: 1 KA, 1 in-situ SCC, and 1(?) case of perforating folliculitis (exons 8 and 5). Moreover 2 missense mutations of TGFBR1 were observed in 1 KA (c.1120G>A; p.Gly374Arg) and in 1 in-situ SCC (c.248C>T; Pro83Leu; Table 1). Those TGBR1 mutations are somatic mutations because they were not present in other samples for the same patient. No additional mutation of the screened exons of BRAF, NRAS, KRAS, AKT1, PI3KCA, PTEN, nor EGFR genes was found in any of the other malignant or benign lesions. Six of these mutations are known to be typically UV induced (Table 1; refs. 13, 14).

Normal skin biopsy specimens

The results of the immunostaining of normal skin samples with Ki-67 and pERK for patients treated with sorafenib or a placebo are shown in Fig. 2.

Figure 2.

Normal skin biopsies of patients treated with sorafenib or placebo. A, Ki-67 immunostaining on normal skin specimen from a placebo-treated patient (left) and a sorafenib-treated patient (right). B, dot graph showing the mean numbers of Ki-67–positive keratinocyte nuclei in 3 representative fields of normal skin samples in placebo- and sorafenib-treated patients. C, numbers and ratios of normal skin specimen expressing p-ERK in the epidermis among the patients treated with placebo or sorafenib for renal cell cancer. D, example of immunostaining results in a placebo-treated patient (left) and a sorafenib-treated patient (right).

Figure 2.

Normal skin biopsies of patients treated with sorafenib or placebo. A, Ki-67 immunostaining on normal skin specimen from a placebo-treated patient (left) and a sorafenib-treated patient (right). B, dot graph showing the mean numbers of Ki-67–positive keratinocyte nuclei in 3 representative fields of normal skin samples in placebo- and sorafenib-treated patients. C, numbers and ratios of normal skin specimen expressing p-ERK in the epidermis among the patients treated with placebo or sorafenib for renal cell cancer. D, example of immunostaining results in a placebo-treated patient (left) and a sorafenib-treated patient (right).

Close modal

Ki-67 staining was found in the nucleus of a few scattered basal cells in nontreated patients whereas it was found in the nucleus of 1 to 2 whole layers or multiple heaps of basal cells in patients treated with sorafenib (Fig. 2A). Ki-67–stained cells were counted by 2 independent pathologists in 3 representative fields of each specimen (15). There was a significant difference between the numbers of Ki-67–positive keratinocytes in the 2 populations of patients with more proliferating cells in the sorafenib-treated group than in the placebo-treated patients (Rank test, P = 0.03; Fig. 2B).

We also found a tendency for increased phosphorylation of ERK in normal skin of sorafenib-treated patients compared with normal skin of placebo-treated patients. A strong positive p-ERK staining was found in 7 out of 11 normal skin samples from sorafenib-treated patients (64%), whereas only 2 out of 8 samples from placebo-treated patients (25%) showed positive, albeit weak and focal p-ERK staining (Fig. 2C and D; P = 0.1, Fisher exact test). The lack of statistical significance might be due here to the small number of specimen.

In vitro results on HaCat keratinocytes

We first used sequencing to verify that HaCat cells did not harbor an oncogenic mutation in any of the RAS genes (HRAS, KRAS, and NRAS; data not shown).

HaCat cells were treated with increasing amounts of sorafenib, and BRAF–CRAF heterodimerization was evaluated by coimmunoprecipitation experiments. We did not detect any heterodimers in DMSO-treated cells, whereas sorafenib induced BRAF–CRAF heterodimers in a dose-dependent manner in keratinocytes (Fig. 3A). As BRAF–CRAF heterodimerization was previously shown to activate CRAF, we measured CRAF kinase activity in keratinocytes treated with increasing doses of sorafenib. Of note, sorafenib is a reversible inhibitor and therefore was no longer present during the in vitro kinase assay. Treatment of HaCat cells with sorafenib stimulated CRAF activity in a dose-dependent manner (Fig. 3B), which paralleled the dimerization of BRAF with CRAF (Fig. 3A), confirming that dimerization activates CRAF in keratinocytes. To analyze the biological effect of sorafenib-induced CRAF activation on the MAPK pathway, we measured ERK phosphorylation in response to sorafenib in a time course experiment. Sorafenib transiently activated ERK at 1, 3, and 10 μmol/L, attaining a maximum after 15 minutes (Fig. 4A). A more pronounced effect was observed with the lower doses of sorafenib. To analyze the biological consequence of ERK activation, we measured the proliferation rate of HaCat cells treated with increasing doses of sorafenib. Cellular proliferation was increased with the low dose (1 μmol/L) of sorafenib. Proliferation was inhibited with higher doses of sorafenib (3 and 10 μmol/L; Fig. 4B).

Figure 3.

Sorafenib induces BRAF–CRAF dimerization and CRAF activation in HaCat keratinocytes. HaCat cells were treated with DMSO (0) and increasing doses of sorafenib (1, 3, or 10 μmol/L) for 4 hours and lysed. A, CRAF was immunoprecipitated and probed for BRAF and CRAF. Quantification is shown below the blot. B, CRAF was immunoprecipitated and its kinase activity against MEK measured in vitro. Similar results were obtained in 3 independent experiments.

Figure 3.

Sorafenib induces BRAF–CRAF dimerization and CRAF activation in HaCat keratinocytes. HaCat cells were treated with DMSO (0) and increasing doses of sorafenib (1, 3, or 10 μmol/L) for 4 hours and lysed. A, CRAF was immunoprecipitated and probed for BRAF and CRAF. Quantification is shown below the blot. B, CRAF was immunoprecipitated and its kinase activity against MEK measured in vitro. Similar results were obtained in 3 independent experiments.

Close modal
Figure 4.

Effect of sorafenib on ERK phosphorylation, cell proliferation, and MCL1 expression in HaCat keratinocytes. A, HaCat cells were treated with DMSO (0) or with increasing doses of sorafenib (1, 3, or 10 μmol/L) for the indicated time and lysed. Lysates were probed for phosphorylated ERK (ppERK) and total ERK. The ratio ppERK/ERK is shown on the graph which represents the mean ± SD of 3 independent experiments. B, proliferation of HaCat cells treated with DMSO (0) an increasing dose of sorafenib for 3 days was measured using the CellTiter assay. The graph represents the mean of 2 experiments in triplicate. C, lysates from (A) were probed for MCL1 and actin. The ratio MCL1/actin was calculated for the indicated dose of sorafenib after 6 or 24 hours of treatment. Graphs represent the mean ± SD of 2 experiments.

Figure 4.

Effect of sorafenib on ERK phosphorylation, cell proliferation, and MCL1 expression in HaCat keratinocytes. A, HaCat cells were treated with DMSO (0) or with increasing doses of sorafenib (1, 3, or 10 μmol/L) for the indicated time and lysed. Lysates were probed for phosphorylated ERK (ppERK) and total ERK. The ratio ppERK/ERK is shown on the graph which represents the mean ± SD of 3 independent experiments. B, proliferation of HaCat cells treated with DMSO (0) an increasing dose of sorafenib for 3 days was measured using the CellTiter assay. The graph represents the mean of 2 experiments in triplicate. C, lysates from (A) were probed for MCL1 and actin. The ratio MCL1/actin was calculated for the indicated dose of sorafenib after 6 or 24 hours of treatment. Graphs represent the mean ± SD of 2 experiments.

Close modal

To determine whether this dual effect on cell proliferation could be explained by the action of sorafenib on the antiapoptotic molecule MCL1, MCL1 expression was studied as a function of sorafenib concentration. We found that MCL1 expression decreased with higher sorafenib concentrations, (3 and 10 μmol/L), whereas 1 μmol/L of sorafenib did not influence MCL1 expression (Fig. 4C). To test whether sorafenib-induced heterodimerization could be detected in situ, we used a PLA. In this assay, a pair of oligonucleotide-labeled secondary antibodies (PLA probes) generates a signal only when they have bound in close proximity, thus allowing the detection of protein–protein interaction in situ. HaCat cells were treated or not with sorafenib and fixed. BRAF–CRAF heterodimerization was visualized by PLA with a combination of a CRAF and a BRAF antibody. Figure 5 shows that sorafenib-induced BRAF–CRAF heterodimerization can be directly detected in keratinocytes.

Figure 5.

PLA showing BRAF–CRAF dimerization induced by sorafenib in keratinocytes in vitro. Sorafenib induces HaCat cells grown on coverslip where treated with DMSO, 1 or 10 μmol/L of sorafenib for 4 hours and fixed. BRAF–CRAF heterodimerization was visualized as red dots by PLA and detected with a fluorescent microcope; cell nuclei were stained with DAPI. Each dot represents 1 BRAF–CRAF complex.

Figure 5.

PLA showing BRAF–CRAF dimerization induced by sorafenib in keratinocytes in vitro. Sorafenib induces HaCat cells grown on coverslip where treated with DMSO, 1 or 10 μmol/L of sorafenib for 4 hours and fixed. BRAF–CRAF heterodimerization was visualized as red dots by PLA and detected with a fluorescent microcope; cell nuclei were stained with DAPI. Each dot represents 1 BRAF–CRAF complex.

Close modal

RNA interference confirmed the specificity of the PLA signal as inhibition of BRAF or CRAF expression with siRNA strongly diminished the PLA signal induced by sorafenib (Supplementary Fig. S1).

Targeted anticancer agents are associated with frequent and diverse cutaneous side effects. The description and study of these skin manifestations is of critical importance. Indeed, they frequently have an adverse impact on the patient's quality of life and understanding their underlying mechanisms is certainly the best way to treat these skin manifestations in addition to offering new perspectives on skin biology (5, 6, 12–14).

Our earlier observations of skin manifestations induced by targeted agents had lead us to formulate the hypothesis that sorafenib-induced KA and skin SCC were due to its effect on RAF kinases. Indeed, these side effects were not seen after blockade of VEGFR, KIT, FLT3, or PDGFR, the other sorafenib targets (5, 12). Our hypothesis was further corroborated by the description of KA and SCC occurring with more specific BRAF inhibitors, vemurafenib and GSK2118436 (2, 15).

In this work, we present a series of 31 skin lesions occurring in 17 patients treated with sorafenib 1 to 9 months after the beginning of the therapy. The lesions were benign and borderline or malignant skin tumors which might seem paradoxic because they occurred while under therapy with an antiproliferative agent.

The entity KA as a distinct tumor at the crossroad between benign and malignant tumors has been debated for decades. KA is a rapidly growing keratotic and crateriform tumor, very similar clinically and pathologically to well-differentiated SCC (16). It is characterized by frequent spontaneous resolution and originates from hair follicles (17). For some authors, it is considered to be low-grade SCC (18). KAs can be observed sporadically, mostly on sun-exposed areas, or in the context of genetic diseases, for example, the multiple self-healing squamous epithelioma (MSSE) also called Ferguson-Smith disease (19). Our pathologic classification between KA and SCC depended on the existence of cytologic atypia, invasive nests or lymphatic metastatic embols. When any of these signs were seen, we classified the crateriform epidermal neoplasms limited by sharp beaks as KA-like SCCs. When neither atypia nor invasion was associated with the crateriform lesion, they were considered as KAs.

The first hypothesis that we put forward following our clinical and pathologic studies of these tumors was that they seemed to belong to a common spectrum of lesions with the possibility of transition from benign lesions, that is, from a cystic more or less inflammatory lesion to KA and SCC. This “continuity” hypothesis is based on several observations. First, several of our patients had associated benign and malignant tumors. All of them had several benign cystic lesions, in addition to the lesions that were biopsied and studied (Table 1). One patient, in particular, developed the complete spectrum of lesions, that is, follicular cysts, perforating folliculitis, KAs, and KA-like SCC (patient # 7). Furthermore, all SCCs were crateriform in shape, resembling KA. We did not observe other clinical subtypes of SCC such as Bowen disease or classic sun-induced, ulcerated, or nodular forms of SCC.

Second, all of these lesions seemed to have developed at the expense of or around hair follicles. Follicular dystrophic lesions and perforating folliculitis are obviously follicular lesions and relics of hair follicles were indeed observed in every biopsy sample. Follicular ontogeny for KA is also well documented (20). The microscopically lamellar structure as well as the type of cytokeratins found in KA suggest that they are hair follicle–derived tumors (21). Thus, our first hypothesis is that all these proliferative epidermal lesions could represent various steps of a same generic process initiated by RAF inhibition in the skin and particularly targeting follicular keratinocytes.

To explore the biological effect of sorafenib on normal skin in vivo, we studied normal skin samples of patients treated with sorafenib or a placebo. Our results showed a significant increase in keratinocyte proliferation and no decrease of ERK phosphorylation, but, on the contrary, a clear tendency to an increase of ERK phosphorylation levels in the skin after treatment with sorafenib. We now have preliminary data on normal skin samples from patients treated with vemurafenib (before treatment and at day 21) showing similar results (data not shown).

These results, which at first glance might seem highly improbable, are in fact in line with recent published data showing that BRAF inhibitors induce activation instead of inhibition of the RAF/MEK/ERK pathway in wtBRAF cells (22–24). The proposed mechanisms underlying the activation of the pathway by a RAF inhibitor imply dimerization of RAF proteins and homodimerization (BRAF–BRAF) or heterodimerization (BRAF–CRAF). Binding of the RAF inhibitor enhances signaling via CRAF engaged in dimerization with BRAF (23). In a second model, binding of the inhibitor induces transactivation of the second element of the homo- or heterodimer (22). In both models, this activation requires wtBRAF together with an additional activation signal of the pathway, as is the case with RAS or EGFR activation. To understand whether this effect could explain keratinocyte proliferation in response to sorafenib, we used HaCat keratinocytes to see whether we could reproduce these results in an in vitro system. We showed that sorafenib indeed regulates the activation of the MAPK pathway and cellular proliferation in keratinocytes in a dose- and time-dependent manner with an increase of both parameters when the dose of sorafenib was low (1 μmol/L). For higher sorafenib concentrations (3 and 10 μmol/L), the effect on ERK phosphorylation was transient and disappeared after 1 hour and the effect on proliferation was rather inhibitory. This is in line with the transactivation model (22) in which RAF inhibitors activate ERK signaling at low concentrations but inhibit it at higher concentrations in wtBRAF cells. The decreased effect of higher concentrations of sorafenib on ERK phosphorylation could be explained by the direct binding and inhibiting activity of the drug on CRAF. As sorafenib has been previously shown to regulate the expression of the antiapoptotic MCL1 molecule (25, 26), we explored the effect of various doses of sorafenib on MCL1 expression. We found that the regulation of MCL1 expression was dependent on sorafenib concentration with inhibited MCL1 expression at higher drug doses. This effect of high doses (3 and 10 μmol/L) of sorafenib on MCL1 expression could explain that, although sorafenib activated ERK at all doses tested, it inhibits cell proliferation at higher doses. Thus, the complex action of sorafenib on the activation of the MAPK kinase pathway and on cell proliferation is dose dependent and results from the combined effects of the drug on RAF kinase inhibition on the one hand and on the control of apoptosis via MCL1 regulation on the other hand. We were able to detect the formation of BRAF–CRAF dimers in vitro by coimmunoprecipitation and directly in cells by PLA for all sorafenib concentrations tested. This dimerization-induced CRAF activation in keratinocytes can explain MAPK activation at least at a low sorafenib concentration which might be the case in the skin. Indeed, the plasma sorafenib concentration in clinical therapeutic use ranges from 3 to 6 mg/L (6.5 to 13 μmol/L) but the drug concentration in the skin is probably much lower than in plasma due to high protein binding (27). In most experimental cell lines models used, RAS activation seems required for RAF dimerization. We show here that RAS is not mutated in the HaCat keratinocytes and is rarely mutated in our biopsy specimen, however, it is possible that RAS is activated by upstream signaling like EGFR activation, which is very frequently observed in the skin. Indeed, we showed that when EGFR activity was inhibited with PD153035 in HaCat cells, sorafenib could not induce RAF heterodimerization anymore (Supplementary Fig. S2). This result shows that activation of the pathway upstream of RAF, presumably including RAS activation, is necessary for sorafenib-induced RAF dimerization in keratinocytes. Our data are, therefore, consistent with models from the literature stating that drug-induced RAF dimerization requires RAS activation either through oncogenic mutation or through RTK activation.

Thus, the model we propose is as follows: BRAF wild-type skin keratinocytes submitted to low sorafenib concentrations activate the MAP kinase pathway via BRAF–CRAF dimerization sparking off CRAF activation which in turn leads to keratinocyte proliferation. Additional events could then precipitate the proliferation and transformation process. In accordance with this model, we found oncogenic mutations (3 HRAS and 3 TP53 mutations and 2 TGFBR1) in benign and malignant skin lesions. Sporadic KA and SC, besides the use of RAF inhibitors, are known to harbor oncogenic RAS and TP53 mutations (30–34). RAS mutations are reported in 30% of KA (HRAS) and in 12 to 46% of 22% of SCC (mostly HRAS; refs. 30–32, 34). Furthermore, TGFBR1 mutations similar to the ones we found in our patients tumors have recently been shown to be germinal mutations directly involved in an autosomal genodermatosis characterized by the occurrence of multiple KAS, the MSSE also known as Ferguson Smith disease (19). Patient 10 had 2 lesions screened for mutations, one KA-like SCC with a TGFBR1 mutation (Pro83Leu) and benign cystic folliculitis that did not harbor any mutation, meaning that the TGFBR1 mutation was somatic and not germinal (Table 1).

The lesions presented by patients with MSSE are clinically and pathologically very similar to lesions presented in the context of RAF inhibitor treatment suggesting an effect of these drugs on the TGFB/TGFBR signaling pathway that warrants further exploration.

Six of the mutations found in our samples are known to be potentially UV induced (patients # 1, 3, 4, 6, 10, and 14), either located on typical UV-sensitive dipyrimidine sites (patients #1, 3, 6, 10, and 14) or on an ACA sequence known to give rise to a mutagenic photoproduct (patient #4; refs. 35, 36). Oncogenic events such as TP53 mutations are found in normal skin keratinocytes on sun-exposed areas (37). Indeed, a recent paper showed that intermittently sun-exposed skin contained an extensive number of intact cells with TP53 mutations, with an estimated annual rate of 35.000 new persistent TP53 mutations per individual (38). In accordance with this hypothesis, the 6 lesions that were harboring mutations with UV signature were located on sun-exposed areas, whereas, the 2 remaining mutations (patients #7 and 12) were found on nonexposed skin areas (palm and buttocks; Table 1). Thus, sorafenib-induced skin tumor might result from the combined effect of paradoxic MAP kinase pathway activation and coexisting or even preexisting oncogenic mutations that could have been generated by UV rays action on normal skin.

Altogether, the fact that we found oncogenic TP53, TGFBR1, and RAS mutations with a UV signature suggests that it is probably a wise move to advise patients receiving sorafenib or other RAF inhibitors to use efficient photoprotective measures.

The fact that we did not find oncogenic mutations in the other malignant lesions could be due to the existence of other mutations that we did not look for or by alternate ways of activating the MAP kinase pathway such as EGFR activation. Indeed, EGFR can also be activated in the skin by various stimuli such as physiologic sun exposure or wound healing (28, 29), and its activation is sufficient to promote RAF dimerization in keratinocytes treated with sorafenib (Supplementary Fig. S2).

It is possible that sun-induced RAF or TP53 mutation or EGFR activation were present in the keratinocyte prior to BRAF inhibition and could have led to keratinocyte transformation in the presence of sorafenib. In some of our samples, a mutational event existing prior to malignant transformation is supported by the fact that we found oncogenic mutations of HRAS and TP53 in 2 benign cystic lesions.

Recently, oncogenic RAS mutations, (mostly HRAS) were found at a higher frequency in 71% of 21 KA and SCC induced by vemurafenib, suggesting that RAS involvement might be more preeminent than other oncogenes in the ontogeny of vemurafenib-induced tumors (39). In conclusion, we propose a model in which paradoxic ERK activation by RAF inhibitors in the skin induces changes in keratinocyte biology with activation of the MAK pathway and cell proliferation associated with a spectrum of skin lesions ranging from more or less inflammatory benign follicular cystic lesions to KA-like SCC. Additional and maybe preexisting somatic genetic events might influence the evolution of the benign lesions to more proliferative and malignant tumors. The fact that these skin lesions occur more frequently with more potent and specific BRAF inhibitors than with the pan-RAF inhibitor sorafenib could result from the differential effect of these drugs on CRAF or from various bioavailabilities of the drugs in the skin. These findings have other important clinical implications. They also suggest that the use of RAF inhibitors might be pointless and even deleterious in normal tissues especially in cells harboring a RAS mutation, as it can be found in lung and colon tissue, for example (40, 41). Indeed, we cannot exclude the hypothesis that RAF inhibitors could promote cell transformation and/or proliferation in these wtBRAF cells with RAS activation and lead to internal tumors. Fortunately, such events have not been reported so far.

Altogether, the effect of RAF inhibitors in the skin illustrates the complexity and the variety of the consequences of the systemic blockade of a universal pathway like the MAPK pathway and reminds us that efficient targeted therapies always have off-target effects that need to be very carefully evaluated and monitored.

C. Robert is participating in advisory boards for Roche and GlaxoSmithKline. J.C. Soria is a consultant for Roche and Bayer. B. Escudier is a consultant for Bayer, Pfizer, GlaxoSmithKline, and Novartis. D. Malka is a consultant for Roche. No potential conflicts of interest were disclosed by the other authors.

The authors thank Lorna Saint Ange for editing.

This work was partly supported by the Institut National du Cancer (INCA 2007-1-PL7) for C. Robert, N. Dumaz, and A. Eychene.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Greenman
C
,
Stephens
P
,
Smith
R
,
Dalgliesh
GL
,
Hunter
C
,
Bignell
G
, et al
Patterns of somatic mutation in human cancer genomes
.
Nature
2007
;
446
:
153
8
.
2.
Flaherty
KT
,
Puzanov
I
,
Kim
KB
,
Ribas
A
,
McArthur
GA
,
Sosman
JA
, et al
Inhibition of mutated, activated BRAF in metastatic melanoma
.
N Engl J Med
2010
;
363
:
809
19
.
3.
Escudier
B
,
Eisen
T
,
Stadler
WM
,
Szczylik
C
,
Oudard
S
,
Siebels
M
, et al
Sorafenib in advanced clear-cell renal-cell carcinoma
.
N Engl J Med
2007
;
356
:
125
34
.
4.
Escudier
B
,
Eisen
T
,
Stadler
WM
,
Szczylik
C
,
Oudard
S
,
Staehler
M
, et al
Sorafenib for treatment of renal cell carcinoma: final efficacy and safety results of the phase III treatment approaches in renal cancer global evaluation trial
.
J Clin Oncol
2009
;
27
:
3312
8
.
5.
Arnault
JP
,
Wechsler
J
,
Escudier
B
,
Spatz
A
,
Tomasic
G
,
Sibaud
V
, et al
Keratoacanthomas and squamous cell carcinomas in patients receiving sorafenib
.
J Clin Oncol
2009
;
27
:
e59
61
.
6.
Autier
J
,
Escudier
B
,
Wechsler
J
,
Spatz
A
,
Robert
C
. 
Prospective study of the cutaneous adverse effects of sorafenib, a novel multikinase inhibitor
.
Arch Dermatol
2008
;
144
:
886
92
.
7.
Hong
DS
,
Reddy
SB
,
Prieto
VG
,
Wright
JJ
,
Tannir
NM
,
Cohen
PR
, et al
Multiple squamous cell carcinomas of the skin after therapy with sorafenib combined with tipifarnib
.
Arch Dermatol
2008
;
144
:
779
82
.
8.
Dubauskas
Z
,
Kunishige
J
,
Prieto
VG
,
Jonasch
E
,
Hwu
P
,
Tannir
NM
. 
Cutaneous squamous cell carcinoma and inflammation of actinic keratoses associated with sorafenib
.
Clin Genitourin Cancer
2009
;
7
:
20
3
.
9.
Kong
HH
,
Cowen
EW
,
Azad
NS
,
Dahut
W
,
Gutierrez
M
,
Turner
ML
. 
Keratoacanthomas associated with sorafenib therapy
.
J Am Acad Dermatol
2007
;
56
:
171
2
.
10.
Robert
C
,
Arnault
J-P
,
Mateus
C
. 
RAF inhibition and induction of cutaneous squamous cell carcinoma
.
Curr Opin Oncol
2011
;
23
:
177
82
.
11.
Dumaz
N
,
Hayward
R
,
Martin
J
,
Ogilvie
L
,
Hedley
D
,
Curtin
JA
, et al
In melanoma, RAS mutations are accompanied by switching signaling from BRAF to CRAF and disrupted cyclic AMP signaling
.
Cancer Res
2006
;
66
:
9483
91
.
12.
Robert
C
,
Soria
JC
,
Spatz
A
,
Le Cesne
A
,
Malka
D
,
Pautier
P
, et al
Cutaneous side-effects of kinase inhibitors and blocking antibodies
.
Lancet Oncol
2005
;
6
:
491
500
.
13.
Robert
C
,
Spatz
A
,
Faivre
S
,
Armand
J-P
,
Raymond
E
. 
Tyrosine kinase inhibition and grey hair
.
Lancet
2003
;
361
:
1056
.
14.
Robert
C
,
Faivre
S
,
Raymond
E
,
Armand
J-P
,
Escudier
B
. 
Subungual splinter hemorrhages: a clinical window to inhibition of vascular endothelial growth factor receptors?
Ann Intern Med
2005
;
143
:
313
4
.
15.
Keeford
R
,
Long
G
,
Arkenau
HT
,
Brown
MP
,
Millward
M
,
O'Day
S
, et al
Selective inhibition of oncogenic BRAF V600E/K/D byGSK2118436: evidence of clinical activity in subjects with metastatic melanoma
.
Pigment Cell Melanoma Res
2010
;
23
:
912
.
16.
Cribier
B
,
Asch
P
,
Grosshans
E
. 
Differentiating squamous cell carcinoma from keratoacanthoma using histopathological criteria. Is it possible? A study of 296 cases
.
Dermatology
1999
;
199
:
208
12
.
17.
Kossard
S
,
Tan
K-B
,
Choy
C
. 
Keratoacanthoma and infundibulocystic squamous cell carcinoma
.
Am J Dermatopathol
2008
;
30
:
127
34
.
18.
Hodak
E
,
Jones
RE
,
Ackerman
AB
. 
Solitary keratoacanthoma is a squamous-cell carcinoma: three examples with metastases
.
Am J Dermatopathol
1993
;
15
:
332
42
;
discussion 343–352
.
19.
Goudie
DR
,
D'Alessandro
M
,
Merriman
B
,
Lee
H
,
Szeverényi
I
,
Avery
S
, et al
Multiple self-healing squamous epithelioma is caused by a disease-specific spectrum of mutations in TGFBR1
.
Nat Genet
2011
;
43
:
365
9
.
20.
Kossard
S
,
Tan
K-B
,
Choy
C
. 
Keratoacanthoma and infundibulocystic squamous cell carcinoma
.
Am J Dermatopathol
2008
;
30
:
127
34
.
21.
Yoshikawa
K
,
Katagata
Y
,
Kondo
S
. 
Relative amounts of keratin 17 are higher than those of keratin 16 in hair-follicle-derived tumors in comparison with nonfollicular epithelial skin tumors
.
J Invest Dermatol
1995
;
104
:
396
400
.
22.
Poulikakos
PI
,
Zhang
C
,
Bollag
G
,
Shokat
KM
,
Rosen
N
. 
RAF inhibitors transactivate RAF dimers and ERK signalling in cells with wild-type BRAF
.
Nature
2010
;
464
:
427
30
.
23.
Heidorn
SJ
,
Milagre
C
,
Whittaker
S
,
Nourry
A
,
Niculescu-Duvas
I
,
Dhomen
N
, et al
Kinase-dead BRAF and oncogenic RAS cooperate to drive tumor progression through CRAF
.
Cell
2010
;
140
:
209
21
.
24.
Hatzivassiliou
G
,
Song
K
,
Yen
I
,
Brandhuber
BJ
,
Anderson
DJ
,
Alvarado
R
, et al
RAF inhibitors prime wild-type RAF to activate the MAPK pathway and enhance growth
.
Nature
2010
;
464
:
431
5
.
25.
Kim
S-H
,
Ricci
MS
,
El-Deiry
WS
. 
Mcl-1: a gateway to TRAIL sensitization
.
Cancer Res
2008
;
68
:
2062
4
.
26.
Ricci
MS
,
Kim
SH
,
Ogi
K
,
Plastaras
JP
,
Ling
J
,
Wang
W
, et al
Reduction of TRAIL-induced Mcl-1 and cIAP2 by c-Myc or sorafenib sensitizes resistant human cancer cells to TRAIL-induced death
.
Cancer Cell
2007
;
12
:
66
80
.
27.
Strumberg
D
,
Voliotis
D
,
Moeller
JG
,
Hilger
RA
,
Richly
H
,
Kredtke
S
, et al
Results of phase I pharmacokinetic and pharmacodynamic studies of the Raf kinase inhibitor BAY 43-9006 in patients with solid tumors
.
Int J Clin Pharmacol Ther
2002
;
40
:
580
1
.
28.
Monslow
J
,
Sato
N
,
Mack
JA
,
Maytin
EV
. 
Wounding-induced synthesis of hyaluronic acid in organotypic epidermal cultures requires the release of heparin-binding egf and activation of the EGFR
.
J Invest Dermatol
2009
;
129
:
2046
58
.
29.
Katiyar
SK
. 
A single physiologic dose of ultraviolet light exposure to human skin in vivo induces phosphorylation of epidermal growth factor receptor
.
Int J Oncol
2001
;
19
:
459
64
.
30.
Pierceall
WE
,
Goldberg
LH
,
Tainsky
MA
,
Mukhopadhyay
T
,
Ananthaswamy
HN
. 
Ras gene mutation and amplification in human nonmelanoma skin cancers
.
Mol Carcinog
1991
;
4
:
196
202
.
31.
Pierceall
WE
,
Mukhopadhyay
T
,
Goldberg
LH
,
Ananthaswamy
HN
. 
Mutations in the p53 tumor suppressor gene in human cutaneous squamous cell carcinomas
.
Mol Carcinog
1991
;
4
:
445
9
.
32.
Spencer
JM
,
Kahn
SM
,
Jiang
W
,
DeLeo
VA
,
Weinstein
IB
. 
Activated ras genes occur in human actinic keratoses, premalignant precursors to squamous cell carcinomas
.
Arch Dermatol
1995
;
131
:
796
800
.
33.
Suk
JD
,
Park
WS
,
Kim
D-K
. 
Low rates of somatic p53 mutations in keratoacanthomas
.
J Dermatol Sci
2009
;
53
:
72
3
.
34.
Corominas
M
,
Kamino
H
,
Leon
J
,
Pellicer
A
. 
Oncogene activation in human benign tumors of the skin (keratoacanthomas): is HRAS involved in differentiation as well as proliferation?
Proc Natl Acad Sci U S A
1989
;
86
:
6372
6
.
35.
Bourre
F
,
Lichtenberger
A
,
Sarasin
A
. 
Comparison of spontaneous and ultraviolet-induced mutagenesis on naked SV40 DNA and SV40 virus
.
Mutat Res
1991
;
250
:
49
53
.
36.
Bourre
F
,
Renault
G
,
Sarasin
A
. 
Sequence effect on alkali-sensitive sites in UV-irradiated SV40 DNA
.
Nucleic Acids Res
1987
;
15
:
8861
75
.
37.
Ling
G
,
Persson
A
,
Berne
B
,
Uhlén
M
,
Lundeberg
J
,
Ponten
F
. 
Persistent p53 mutations in single cells from normal human skin
.
Am J Pathol
2001
;
159
:
1247
53
.
38.
Ståhl
PL
,
Stranneheim
H
,
Asplund
A
,
Berglund
L
,
Pontén
F
,
Lundeberg
J
. 
Sun-induced nonsynonymous p53 mutations are extensively accumulated and tolerated in normal appearing human skin
.
J Invest Dermatol
2011
;
131
:
504
8
.
39.
Lacouture
ME
,
Chapman
PB
,
Ribas
A
,
Sosman
JA
,
Mc Arthur
GA
,
Flaherty
K
, et al
Presence of frequent underlying RAS mutations in cutaneous squamous cell carcinomas and keratoacanthomas (cuSCC/KA) that develop in patients during vemurafenib therapy
.
J Clin Oncol
2011
;
29
:
8520
.
40.
Parsons
BL
,
Marchant-Miros
KE
,
Delongchamp
RR
,
Verkler
TL
,
Patterson
TA
,
McKinzie
PB
, et al
ACB-PCR quantification of K-RAS codon 12 GAT and GTT mutant fraction in colon tumor and non-tumor tissue
.
Cancer Invest
2010
;
28
:
364
75
.
41.
Keohavong
P
,
Mady
HH
,
Gao
WM
,
Siegfried
JM
,
Luketich
JD
,
Melhem
MF
. 
Topographic analysis of K- ras mutations in histologically normal lung tissues and tumours of lung cancer patients
.
Br J Cancer
2001
;
85
:
235
41
.