Melanocytic nevi are benign proliferations of pigment cells that can occasionally develop into melanomas. There is a significant correlation between increased nevus numbers and melanoma development. Our previous reports revealed that 7,12-dimethylbenz(a)anthracene (DMBA) and 12-O-tetradecanoyl-phorbol-13-acetate (TPA) induced dysplastic nevi in C3H/HeN mice, with a potential to transform into melanomas. To understand the immune mechanisms behind this transformation, we applied increasing DMBA doses followed by TPA to the skin of C3H/HeN mice. We observed that increased doses of DMBA correlated well with increased numbers of nevi. The increased DMBA dose induced diminished immune responses and promoted the expansion of regulatory T cells (Treg) that resulted in increased IL10 and reduced IFNγ levels. Mice with increased nevus numbers had loss of p16 expression. These mice had increased migration of melanocytic cells to lymph nodes (LN) and a greater percent of LNs produced immortalized melanocytic cell lines. DMBA-induced immunosuppression was lost in CD4-knockout (KO) mice. Lymphocytes in the CD4KO mice produced less IL10 than CD8KO mice. Furthermore, CD4KO mice had significantly reduced nevus numbers and size compared with wild-type and CD8KO mice. These results suggest that Tregs play a vital role in the incidence of nevi and their progression to melanoma.

Prevention Relevance: There has been little progress in developing novel strategies for preventing premalignant dysplastic nevi from becoming melanomas. In this study in mice, regulatory-T cells enhanced progression of benign nevi to malignant melanomas; and by inhibiting their activity, melanomas could be retarded. The findings identify new possibilities for melanoma prevention in high risk individuals.

Melanocytic nevi are abnormal clusters of pigment-producing cells that accumulate in the dermis and epidermis. Although they begin as benign lesions, and most remain so, a small proportion progresses to become malignant melanomas. The risk of melanoma varies based on type, size, number, and location of nevi. In approximately 4%–25% of nonfamilial cases, melanoma occurs in conjunction with a preexisting nevus (1). Patients with familial nevus syndrome have increased numbers of nevi that are dysplastic in nature. They have almost 100-times higher risk of developing melanoma than the general population, and approximately 50% will develop at least one melanoma by the age of 50 years (2). Another situation in which individuals develop increased numbers of nevi is in eruptive melanocytic nevi (EMN). EMN are characterized by the quick appearance of large numbers (>100) of nevi, often in a grouped distribution. EMN are associated with systemic immunosuppression, such as that seen after organ transplantation (3, 4). The paucity of EMN cases reported in the literature has made it difficult to assess a correlation between EMN and an increased risk for melanoma (3). However, sequencing of EMN revealed the presence of BRAFV600E mutations in 85% of lesions (5). The role of mutated BRAF in the initiation and progression of melanoma is well known and suggests there may be a correlation between immunosuppression, EMN, and melanoma development.

Basic research and clinical observations over the past several decades have clearly demonstrated that immunologic mechanisms are capable of controlling melanoma growth. This line of investigation has led to the introduction of novel immunotherapeutic agents that prolong the survival of patients with advanced melanomas (6–9). Most melanomas begin as premalignant dysplastic nevi, which after months to years, may progress to become invasive melanomas. Thus, there is ample opportunity to prevent dysplastic nevi from evolving into melanomas. Identification of novel preventive agents that can avert melanoma development has proceeded slowly, at least, in part, because of the limited number of preclinical models that can be used to evaluate potential protective modalities. This is particularly true for immunologic interventions that could prevent dysplastic nevi from becoming melanomas. Existing mouse melanoma models are largely restricted to transplantable syngeneic melanoma lines, such as B16, compatible only with C57BL/6 mice, or transgenic mice with enforced expression of mutant oncogenes. These animal models rapidly develop melanomas without first showing evidence of dysplastic nevi. Moreover, it is difficult to study immune surveillance because the genes are expressed in all tissue-specific cells (defined by promoter for transgene).

Our previous work (10) has shown that a single topical application of 7,12-dimethylbenz(a)anthracene (DMBA) followed by weekly application of 12-O-tetradecanoyl-phorbol-13-acetate (TPA) induces melanocytic nevi in C3H/HeN mice. In this model, mice develop large numbers of pigmented nevi, some of which progress to become invasive melanomas and then metastasize to regional lymph nodes (LN; ref. 10). Many of the clinical, genetic, and biochemical features closely resemble those that occur in humans. This model gives us flexibility to study the role of immune mediators in melanoma, as well as to understand its initiating and promoting mechanisms.

In this study, we observed that increased DMBA doses correlated well with increased nevus numbers, and were also responsible for expansion of regulatory T cells. Regulatory T cells (Treg) were essential for immunosuppression and nevus development.

Animals

Male and female C3H/HeN mice ages 6–8 weeks, obtained from Charles River Laboratories, were housed in the University of Alabama at Birmingham (UAB, Birmingham, AL) pathogen-free animal facility. These mice were bred and female C3H/HeN mice so obtained were used for carcinogenesis experiments, beginning at 8–10 weeks of age. For other experiments, a mix of male or female animals was used. Each group had the same number of age-matched male or female mice. All animals were fed a normal diet (standard chow) and were given water ad libitum. The UAB Institutional Animal Care and Use Committee (Birmingham, AL) approved the animal protocol for the study (approval# 101009248). CD4- and CD8-knockout (KO) mice were backcrossed onto the C3H/HeN background and used as described in our earlier studies (11).

Chemicals and antibodies

DMBA (≥95% purity), 1-fluoro-2,4-dinitrobenzene (DNFB), N6, 2′-O-dibutyryladenosine 3:5-cyclic monophosphate (dbcAMP), and sodium orthovanadate (Na3VO4) were purchased from Sigma-Aldrich Chemical Co. TPA was obtained from LC Laboratories. CD4-PE (RRID:AB_465506), CD4-FITC (AB_464892), FOXP3-PE (AB_465936), IL-10-PE (AB_466176), CD45.2-Percp-Cy5.5 (AB_953590), and CD45.2-FITC (AB_465061) were obtained from eBioscience. FOXP3-v450 (AB_10611728), IFNγ-PE-Cy7 (AB_396766), CD8-Alexa-647 (AB_396792), and CD8-PE (AB_394571) were obtained from BD Pharmingen.

Nevus/melanoma induction protocol

The DMBA/TPA nevogenesis protocol was applied as described previously (10). The shaved and naired backs of mice were painted with increasing DMBA doses [100–1,000 μg (∼400–4,000 nanomoles (nmoles))] of DMBA in 100 μL of acetone]. The animals were then treated biweekly with 12.5 μg TPA (20 nmol) in acetone. Lesional area was measured by using length and width of all nevi and using the following formula to calculate the area of an ellipse: [area = π (d1/2)(d2/2)]. All mice were naired weekly to count and measure the nevi.

Development of melanocytic cell lines from LNs

LNs were digested in 200 μL of digestion buffer [collagenase D (Roche; 1 mg/mL) and DNase (20 μg/mL)] for 45 minutes. The cells were cultured in melanocyte growth media [OptiMEM with dbcAMP [0.1 millimolar (mmol)], Na3VO4 (1 μmol/L), horse serum (7%), and TPA (25 ng/mL)]. Medium was changed every 2 days.

Contact hypersensitivity response and adoptive transfer

Mice were sensitized (initiation) on the abdomen with 100 μL of DMBA (0.1%–1% w/v DMBA in acetone). Elicitation of specific responses was assessed 5 days after DMBA sensitization. DMBA (20 μL of 0.1%–1% w/v DMBA in acetone) was painted on the ears for elicitation. Baseline ear thickness measurements were taken before elicitation and further measurements were taken after elicitation daily for the next 5 days using a dial thickness gauge Spring-loaded Micrometer (Mitutoyo 7301). The maximum increment in ear thickness compared with the baseline preelicitation level was used to quantify the net ear thickness. Naïve mice, which were not sensitized, but were challenged with DMBA, served as negative controls. To assess the extent of suppression by DMBA-specific regulatory T cells of each group, mice were sensitized as described. Single-cell suspensions of LN cells were obtained after 5 days. CD4+ T cells were isolated by Magnetic Bead Separation (Miltenyi Biotec) according to the manufacturer's instructions. Then, 10 × 106 CD4+ T cells were adoptively transferred into wild-type (WT) mice. After 24 hours, mice were sensitized with DMBA as described. Elicitation and assessment of ear thickness were performed as described above.

Flow cytometry

Ear skin or LNs from DMBA/TPA-treated mice or age-matched untreated skin samples were collected as described previously (10). Ears from at least 3 mice were split in ventral and dorsal side, minced with scissors, and digested with collagenase D/DNase for 1 hour. Cells were passed through a 70-μm screen, counted, and used for staining with indicated antibodies. Single-cell suspensions were prepared individually by collagenase D/DNase I digestion. LN cells were also prepared using collagenase digestion. Cells were collected, counted, and dispensed at 1 × 106 cells per sample and pretreated with Fc receptor block, and then stained with the appropriate primary antibody. After fixation/permeabilization, cells were further stained with the appropriate antibodies and fluorochromes. Flow cytometric analysis was performed using a BD LSRII cytometer with BD FACS Diva Software for acquisition and FlowJo 9.5.2 for data analysis (10).

T-cell cultures and cytokine assay

For analysis of cytokines and T-cell subsets during contact hypersensitivity (CHS), mice were sacrificed on day 3 after DMBA challenge. Ear draining LNs were removed and minced with scissors, and digested in Hank's Balanced Salt Solution containing collagenase D (1 mg/mL, Roche Applied Sciences) and 20 μg/mL DNAse I (Sigma) for 45 minutes. After cells were passed through 100-μm mesh, they were counted and 2 × 106 cells per mouse were stimulated with PMA (50 ng/mL) and ionomycin (250 ng/mL) for 5 hours in the presence of Brefeldin A (2 μmol/L) for intracellular staining of cytokines. LNs from naïve mice were processed in parallel as a negative control. Staining profiles were obtained using LSRII Flow Cytometer (BD Biosciences) and FlowJo v9.5.2 for Mac or v10.0 for Windows computers.

RNA extraction and RT-PCR

The total genomic DNA and RNA were extracted from samples using TRizol Reagent (Invitrogen) according to the manufacturer's instructions. The concentration of total RNA was determined by measuring the absorbance at 260 nm using an Eppendorf biophotometer plus. Purity of isolated RNA was determined with the ratio of absorbance 260/280 nm > 1.8. cDNA was synthesized from 1 μg RNA using a Reverse Transcriptase Kit (Bio-Rad) according to the manufacturer's instructions. The expression of p16INK4a and p19ARF mRNA was detected using primers and conditions described elsewhere (12). For Gapdh primers were, forward: 5′-CATGTTCCAGTATGACTCCACTC-3′ and reverse: 5′- GGCCTCACCCCATTTGATGT-3′ (13). The primers used for H-ras were WT forward: 5′-CAGCAGGTCAAGAAGAGTATAGTGCCA-PO4–3; mutant forward: 5′-CATCTTAGACACAGCAGGTCT-3′; and common reverse: 5′-GCGAGCAGCCAGGTCACAC-3′. The blocker allele- and mutant allele–specific primers were used at a 4:1 ratio (WT:mutant) using RT-PCR protocols that were optimized for increased specificity and sensitivity in detecting the H-ras Q61L point mutation. Thermocycling conditions were 1 minute at 95°C and 35 cycles of 20 seconds at 95°C, 30 seconds at 60°C, and 20 seconds at 72°C. We followed the established allele-specific competitive blocker PCR as per Parsons and colleagues (14), the WT allele–blocking primer was phosphorylated at the 3′ end to block primer extension.

Statistical analysis

Data were analyzed with GraphPad Prism 7.0 software. One-way ANOVA was performed to determine statistical significance. For correlations, equal distribution was assessed and linear regression was calculated and reported as R2. Statistical significance was set at a P < 0.05.

Larger doses of DMBA used for immunization decrease the DMBA allergic CHS response

We sensitized mice with increasing DMBA doses [100 μL of 0.1% (400 nmol), 0.5% (2,000 nmol), and 1% (4,000 nmol)] and analyzed the allergic CHS responses to DMBA by challenging with 20 μL of 0.1% (80 nmol) DMBA on the ears. We observed that there was a dose-dependent decrease in ear swelling responses in the mice sensitized with larger DMBA doses (Fig. 1A). Contrary to the reduction in CHS when higher amounts of DMBA were given for sensitization, increasing the dose at elicitation produced a dose-dependent increase in the ear swelling response (Fig. 1B). Next, we analyzed whether larger DMBA doses reduced the allergic CHS response to unrelated haptens as well. We painted mice with DMBA and 24 hours later applied DNFB at the same site. After challenging with DNFB on day 5 and measuring the ear swelling response, we observed that there was a dose-dependent reduction in ear swelling as DMBA doses increased (Fig. 1C). We next analyzed the effect of DMBA dose mutations in the skin. Mice were sensitized with DMBA on the abdomen and were treated 5 days later with 0.1% DMBA on the back. Five days after that, back skin was collected for mRNA analysis of mutant H-ras. We observed that mice immunized with larger DMBA doses showed greater numbers of H-ras mutations even when examined in the skin at a distant site (Fig. 1D).

Figure 1.

The magnitude of the DMBA-induced immune response is dependent on the dose at sensitization and not challenge. A, Mice were sensitized on the abdomen with increasing doses of DMBA (100 μL of 0.1%, 0.5%, or 1%) and challenged 5 days later on the ears with 2 μL of 0.1% DMBA. B, Mice were sensitized on the abdomen with DMBA (100 μL of 0.1%) and challenged 5 days later on the ears with increasing doses of DMBA (20 μL of 0.1%, 0.5%, or 1%). C, Mice were sensitized on the abdomen with increasing doses of DMBA (100 μL of 0.1%, 0.5%, and 1%). Twenty-four hours later, mice were resensitized with 25 μL of 0.3% (v/v) DNFB. Five days later, mice were challenged with 10 μL of 0.2% (v/v) DNFB on the ear. There were 4–6 mice per group. D, Relative mutant H-ras mRNA expression 5 days after DMBA application. Mice were sensitized with the indicated DMBA doses on the abdomen and challenged on the back 5 days later with 0.1% DMBA. Five days later, skin was collected for mRNA analysis of mutant H-ras as indicated in the figure. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ns, not significant. There were 2–3 mice per group and the experiment was done twice.

Figure 1.

The magnitude of the DMBA-induced immune response is dependent on the dose at sensitization and not challenge. A, Mice were sensitized on the abdomen with increasing doses of DMBA (100 μL of 0.1%, 0.5%, or 1%) and challenged 5 days later on the ears with 2 μL of 0.1% DMBA. B, Mice were sensitized on the abdomen with DMBA (100 μL of 0.1%) and challenged 5 days later on the ears with increasing doses of DMBA (20 μL of 0.1%, 0.5%, or 1%). C, Mice were sensitized on the abdomen with increasing doses of DMBA (100 μL of 0.1%, 0.5%, and 1%). Twenty-four hours later, mice were resensitized with 25 μL of 0.3% (v/v) DNFB. Five days later, mice were challenged with 10 μL of 0.2% (v/v) DNFB on the ear. There were 4–6 mice per group. D, Relative mutant H-ras mRNA expression 5 days after DMBA application. Mice were sensitized with the indicated DMBA doses on the abdomen and challenged on the back 5 days later with 0.1% DMBA. Five days later, skin was collected for mRNA analysis of mutant H-ras as indicated in the figure. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ns, not significant. There were 2–3 mice per group and the experiment was done twice.

Close modal

Increased DMBA doses enhance nevus incidence and size

Because higher DMBA doses at immunization resulted in a reduced DMBA allergic CHS response, we next analyzed whether increased dose of DMBA augmented the number and growth of nevi. We observed that the nevus size and numbers per mouse increased with a large DMBA dose (Fig. 2A and B). Furthermore, the average size of individual nevi also increased as the dose of DMBA increased (Fig. 2C). Categorizing the nevi in different sizes also revealed that nevi that were greater in size were more prevalent in mice that received a large DMBA dose compared with mice that received a lower DMBA dose (Fig. 2D). The nevus numbers and DMBA doses correlated significantly (P = 0.001; Fig. 3A). Furthermore, the percent loss of p16INK4a expression in lesions correlated well with nevus numbers (P = 0.01; Fig. 3B). The percent LNs that produced an immortalized cell line also correlated significantly with average nevus numbers (P = 0.0068; Fig. 3C).

Figure 2.

A larger DMBA dose enhances the incidence and size of nevi. Mice were painted on the backs with increasing doses of DMBA (100 μL of 0.1%, 0.5%, and 1%) and promoted with 12.5 μg TPA beginning 1 week later. Number of nevi per mouse (A), nevus area per mouse (B), average nevus size (C), and percent distribution of lesions (D), 15 weeks after initiation of the nevogenesis protocol. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ns, not significant. There were at least 8–10 mice per group.

Figure 2.

A larger DMBA dose enhances the incidence and size of nevi. Mice were painted on the backs with increasing doses of DMBA (100 μL of 0.1%, 0.5%, and 1%) and promoted with 12.5 μg TPA beginning 1 week later. Number of nevi per mouse (A), nevus area per mouse (B), average nevus size (C), and percent distribution of lesions (D), 15 weeks after initiation of the nevogenesis protocol. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ns, not significant. There were at least 8–10 mice per group.

Close modal
Figure 3.

The number of nevi correlates with DMBA dose and aggressiveness of melanoma progression. A, Increasing DMBA doses correlates well with nevus incidence. B, Loss of p16 in the lesions correlates well with the number of nevi present on the mice. C, Increased nevus number correlates with increased ability to generate immortalized melanoma cells from LNs. There were at 8–10 mice per group. Linear regression analysis was performed as described in Materials and Methods and R2 and P values are shown in each figure.

Figure 3.

The number of nevi correlates with DMBA dose and aggressiveness of melanoma progression. A, Increasing DMBA doses correlates well with nevus incidence. B, Loss of p16 in the lesions correlates well with the number of nevi present on the mice. C, Increased nevus number correlates with increased ability to generate immortalized melanoma cells from LNs. There were at 8–10 mice per group. Linear regression analysis was performed as described in Materials and Methods and R2 and P values are shown in each figure.

Close modal

Treg expansion and function are increased by increasing the DMBA dose

In prior studies, we have shown that in C3H/HeN mice, CD8+ T cells are the major effector cells for DMBA CHS; in contrast, CD4+ T cells have a regulatory function (11). We next investigated the nature of T cells in draining LNs after DMBA-induced CHS. We observed a minor increase in frequency of CD4+ T cells and a minor decrease in frequency of CD8+ T cells as the DMBA dose increased (Fig. 4A). However, there was a significant increase in frequency of CD25+Foxp3+CD4+ T cells as the DMBA dose increased (Fig. 4A). This was accompanied by a 10-fold increase in IL10 levels and a 50% decrease in IFNγ levels at highest dose compared with the lowest DMBA dose (Fig. 4B). Because immune responses at the site of exposure are also important, we next analyzed T cells in the ears. We observed a reduction in CD8+Ki67+ T cells in the LNs of the mice at the higher dose, indicating that the proliferation of CD8+ T cells is impaired or suppressed after a DMBA dose increase. Like the draining lymph nodes (dLNs), we observed an increase in CD4+ and decrease in CD8+ T-cell infiltration in the ear skin (Fig. 4C). To confirm that the regulatory CD4+ T cells mediate DMBA-induced immunosuppression, we applied increasing DMBA doses to CD4KO mice. We observed that CD4KO mice were resistant to DMBA-induced immunosuppression compared with WT mice (Fig. 4D). Consistent with our previous observations (11), we found an exaggerated DMBA CHS in CD4KO mice compared with WT mice (Fig. 4D). Transfer of CD4+ T cells from mice that were sensitized with increasing doses of DMBA revealed that these cells showed an enhanced suppressive phenotype compared with nonspecific and CD4+ T cells from low-dose DMBA-treated mice (Fig. 4E).

Figure 4.

Increased Treg expansion and function with increased DMBA dose in skin. A and B, Mice were sensitized on the abdomen with increasing doses of DMBA (100 μL of 0.1%, 0.5%, and 1%) and challenged 5 days later on ears with 20 μL of 0.1% DMBA. Ear draining LNs were isolated 3 days after DMBA challenge and stained with anti-CD4, CD8, CD25, and Foxp3 antibodies and analyzed by flow cytometry. A total of 1 × 106 cells from each group were stimulated with PMA/ionomycin for 48 hours and supernatants were analyzed for IL10 and IFNγ by ELISA. Histograms are four LNs pooled from 2 mice and experiments were done twice. C, Mice were sensitized and challenged with DMBA as above. Three days after sensitization, ears were digested with collagenase D/DNase as described in the Materials and Methods. Cells were gated on CD45.2 as shown. Cells were stained for markers as shown in the figure. The ears from 3 mice were pooled and digested to release the cells. D, WT and CD4KO mice were sensitized with increasing DMBA doses and challenged as described in A and B. E, Mice were sensitized with increasing DMBA doses, and 5 days later CD4 T cells were collected from each group and injected into WT mice. Twenty-four hours after the adoptive transfer, mice were sensitized with DMBA on the abdomen and challenged on the ears 5 days later. Each group had 4–5 mice per group (*, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001; ns, not significant).

Figure 4.

Increased Treg expansion and function with increased DMBA dose in skin. A and B, Mice were sensitized on the abdomen with increasing doses of DMBA (100 μL of 0.1%, 0.5%, and 1%) and challenged 5 days later on ears with 20 μL of 0.1% DMBA. Ear draining LNs were isolated 3 days after DMBA challenge and stained with anti-CD4, CD8, CD25, and Foxp3 antibodies and analyzed by flow cytometry. A total of 1 × 106 cells from each group were stimulated with PMA/ionomycin for 48 hours and supernatants were analyzed for IL10 and IFNγ by ELISA. Histograms are four LNs pooled from 2 mice and experiments were done twice. C, Mice were sensitized and challenged with DMBA as above. Three days after sensitization, ears were digested with collagenase D/DNase as described in the Materials and Methods. Cells were gated on CD45.2 as shown. Cells were stained for markers as shown in the figure. The ears from 3 mice were pooled and digested to release the cells. D, WT and CD4KO mice were sensitized with increasing DMBA doses and challenged as described in A and B. E, Mice were sensitized with increasing DMBA doses, and 5 days later CD4 T cells were collected from each group and injected into WT mice. Twenty-four hours after the adoptive transfer, mice were sensitized with DMBA on the abdomen and challenged on the ears 5 days later. Each group had 4–5 mice per group (*, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001; ns, not significant).

Close modal

Number and size of nevi are dependent on CD4 T cells

Because we observed an important role for regulatory T cells in the magnitude of DMBA-induced immune response, WT, CD4KO, and CD8KO mice were subjected to our nevogenesis protocol to analyze the incidence and size of pigmented lesions. We observed that CD4KO mice were resistant to increases in the number and area of nevi compared with WT and CD8KO mice (Fig. 5A and B). Furthermore, the average size of pigmented lesions was also significantly reduced in CD4KO mice compared with WT and CD8KO mice. However, we also observed a slight increase in average size of lesions in CD8KO mice compared with WT mice, indicating that CD8 T cells control the growth of the lesions vis-à-vis incidence (Fig. 5C). Categorizing the nevi in different size groups also revealed that smaller sized nevi were more prevalent in CD4KO mice compared with WT and CD8KO mice (Fig. 5D). The DMBA-induced CHS responses, as described earlier, were exaggerated in CD4KO and were significantly diminished in CD8KO compared with WT mice (Fig. 5E). The percent lesions that produced a cell line were also greater in CD8KO (35%) and WT (25%) compared with CD4KO (10%), suggesting a more benign behavior of lesions obtained from CD4KO mice.

Figure 5.

Reduced number of pigmented lesions in CD4KO compared with WT and CD8KO mice. DMBA (100 μg/mouse) was applied on the shaved and naired backs of mice and 1 week later TPA (12.5 μg/mouse) was applied twice weekly for 25 weeks. Nevi were counted and the area was measured weekly. A, WT and CD8KO mice had nearly equal lesion numbers and these numbers were significantly greater than CD4KO mice. B, Reduced lesion area per mouse was observed in CD4KO mice, but not in CD8KO or WT mice. Like the lesion numbers, the area was significantly reduced in CD4KO mice as compared with WT and CD8KO mice. The lesion area per mouse between CD8KO and WT groups was equal. C, Area of each individual lesion at week 25 for each group was measured. The mean lesion area was greatly reduced in CD4KO mice as compared with both WT and CD8KO. CD8KO mice had lesion whose size was slightly larger than WT mice. D, Stacked bar graph for lesion area at week 25 from each group, shows that in CD8KO mice there is a higher percentage of lesions greater than 3 or 6 mm2 as compared with WT mice and CD4KO mice have larger percentage in the range below 3 mm2. E, Day 3 allergic CHS responses in CD4KO mice were increased significantly compared with WT and CD8KO mice. Each group had 8–10 mice per group (*, P < 0.05; **, P < 0.01; ***, P < 0.001).

Figure 5.

Reduced number of pigmented lesions in CD4KO compared with WT and CD8KO mice. DMBA (100 μg/mouse) was applied on the shaved and naired backs of mice and 1 week later TPA (12.5 μg/mouse) was applied twice weekly for 25 weeks. Nevi were counted and the area was measured weekly. A, WT and CD8KO mice had nearly equal lesion numbers and these numbers were significantly greater than CD4KO mice. B, Reduced lesion area per mouse was observed in CD4KO mice, but not in CD8KO or WT mice. Like the lesion numbers, the area was significantly reduced in CD4KO mice as compared with WT and CD8KO mice. The lesion area per mouse between CD8KO and WT groups was equal. C, Area of each individual lesion at week 25 for each group was measured. The mean lesion area was greatly reduced in CD4KO mice as compared with both WT and CD8KO. CD8KO mice had lesion whose size was slightly larger than WT mice. D, Stacked bar graph for lesion area at week 25 from each group, shows that in CD8KO mice there is a higher percentage of lesions greater than 3 or 6 mm2 as compared with WT mice and CD4KO mice have larger percentage in the range below 3 mm2. E, Day 3 allergic CHS responses in CD4KO mice were increased significantly compared with WT and CD8KO mice. Each group had 8–10 mice per group (*, P < 0.05; **, P < 0.01; ***, P < 0.001).

Close modal

Immunosuppressive microenvironment created by CD4 cells

Because CD4 cells promoted nevus incidence and growth, we next investigated the cytokines being produced by the CD4KO mice. As CD4KO lack CD4 cells, the CD8+ cells compensated for the loss, and similarly CD4+ cells compensated for the loss in CD8KO (Fig. 6A). The percent of CD25+Foxp3+ cells gated on CD4 was similar, but the overall percentage of Tregs per LN was greater in CD8KO mice (11% vs. 15%). The percentage of CD8+Foxp3+ cells was similar in CD4KO and WT mice. There was increased IL10 production from CD4 cells of CD8KO mice compared with the CD4+ cells of WT mice. Similarly, the CD8+ T cells from CD4KO mice produced increased IFNγ compared with CD8+ T cells from WT mice. We confirmed IL10 and IFNγ production using ELISA and observed IL10 levels were increased in CD8KO and WT mice and IFNγ was mainly produced by CD8+ T cells (Fig. 6B). Investigation of infiltrating cells revealed that a reduced number of CD45.2 cells infiltrated the skin in CD8KO mice compared with WT or CD4KO mice. The CD4+Foxp3+ cells were the major cell type that was present in the skin after DMBA application and CD8+ T cells were major IFNγ producers (Fig. 6C).

Figure 6.

CD4 T cells are the major source of IL10. A, Mice were sensitized on the abdomen with DMBA (100 μL of 0.1%) and challenged 5 days later on ears with 20 μL of 0.1% DMBA. Ear draining LNs were isolated 3 days after DMBA challenge and stained with anti-CD4, CD8, CD25, and Foxp3 antibodies and analyzed by flow cytometry. B, A total of 1 × 106 cells from each group were stimulated with PMA/ionomycin for 6 and 48 hours for intracellular staining and ELISA, respectively, to detect IL10 and IFNγ. Histograms are from four LNs pooled from 2 mice. The experiment was done twice. C, Mice were sensitized and challenged as above. Three days after sensitization, ears were digested with collagenase D/DNAse as described in Materials and Methods. Cells were gated on CD45.2 as shown. Cells were stained for the markers as shown in the figure. The ears from 3 mice were pooled and digested to release the cells (**, P < 0.01).

Figure 6.

CD4 T cells are the major source of IL10. A, Mice were sensitized on the abdomen with DMBA (100 μL of 0.1%) and challenged 5 days later on ears with 20 μL of 0.1% DMBA. Ear draining LNs were isolated 3 days after DMBA challenge and stained with anti-CD4, CD8, CD25, and Foxp3 antibodies and analyzed by flow cytometry. B, A total of 1 × 106 cells from each group were stimulated with PMA/ionomycin for 6 and 48 hours for intracellular staining and ELISA, respectively, to detect IL10 and IFNγ. Histograms are from four LNs pooled from 2 mice. The experiment was done twice. C, Mice were sensitized and challenged as above. Three days after sensitization, ears were digested with collagenase D/DNAse as described in Materials and Methods. Cells were gated on CD45.2 as shown. Cells were stained for the markers as shown in the figure. The ears from 3 mice were pooled and digested to release the cells (**, P < 0.01).

Close modal

Melanocytic nevi can appear in both sun-exposed and -unexposed skin. Their etiology includes genetic factors, excessive exposure to sunlight, and other environmental influences, such as pollutants in the environment. An analysis of the scientific literature reveals many correlations between polyaromatic hydrocarbon exposure and the occurrence of malignant melanoma, particularly on cutaneous surfaces of skin that are protected from sunlight (15). The polyaromatic hydrocarbon, DMBA, is a model carcinogen that induces epidermal malignancies of skin, and a major focus of study has been its role in the initiation of nonmelanoma skin cancers (16). Our previous work demonstrated that a single dose of 400 nmol of DMBA followed by weekly application of TPA resulted in melanocytic nevi in C3H/HeN mice that have the potential to progress to melanomas. This model gave us the opportunity to study the involvement of immune mediators on earlier stages of melanoma development, rather than on melanomas already present. This is an important issue, whether the immune system is to be manipulated to prevent melanomas in high-risk individuals. In this study, we report for the first time that CD4+ T cells promote initiation and growth of melanocytic nevi.

Our previous studies had shown that the generation of cytotoxic T cells is, in part, responsible for limiting the development of the melanocytic nevi (17). During the course of these experiments, we observed that application of larger doses of DMBA produced greater numbers of nevi that grew to a larger size. While it might be expected that greater amounts of DMBA would produce more and larger sized pigmented lesions, we found this occurred even at a distant skin site. Moreover, the larger doses of DMBA resulted in a smaller CHS response than lower doses. We, therefore, postulated that an increased number of Tregs might contribute to the suppressed DMBA CHS response and to the greater numbers and larger size of the melanocytic lesions.

Tregs suppress a variety of physiologic and pathologic immune responses (18) and play a key role in progression of various cancers. They directly promote immunosuppression by regulating effector T-cell functions (19). They can also directly eliminate effector T cells and compete for dendritic cells (DC) for antigen presentation. Furthermore, they are known to suppress DC polarization (20). Immunosuppression in the melanoma microenvironment is facilitated by Tregs and is a major mechanism of immune escape once melanomas are already present. Tregs accumulate in melanoma and the reduced ratio of CD8 to Tregs is used as a predictive marker for survival of patients with melanoma (21). Immunotherapies using neutralizing antibodies to cell surface markers of Tregs, such as CTLA-4, have been promising, especially in melanoma immunotherapy (6–9). Although there is an evidence of a Treg contribution to melanoma progression, little was known about their role in melanoma precursors.

We found, that employing our murine model for nevus initiation and progression by increasing the dose of DMBA, there was an expansion of functional CD4+ regulatory T cells. These cells were essential for both suppression of DMBA CHS and the increase in nevi that develop. We also found that an increased percentage of LNs produced immortalized cell lines from mice that had fewer Tregs, indicating an association between immunosuppression and melanocyte immortalization in LNs.

Previous studies in mice have demonstrated that repeated DMBA doses over time have a greater carcinogenic affect than the same amount administered as a total single dose of DMBA (22). Repeated hapten exposure can induce a Treg component and hence, promote tumorigenesis. Others and our group has previously demonstrated that CD4+ T cells, presumably Tregs, promote squamous cell carcinoma tumor development in DMBA-, ultraviolet (UV)-, and virus-induced tumor models. In all cases, CD4+ T cells enhanced the development of tumors (11, 13, 23).

Previously, we have described the cytokine profile of CD8+ and CD4+ T cells after cutaneous application of DMBA on C3H/HeN mice (11). In this study, we confirmed our previous results that CD4+ T cells are the main cells that secrete IL10, while CD8+ cells are the IFNγ producers. Specifically, infiltration of cells after DMBA challenge was impaired in CD8KO mice. Loss of CD8+ T cells did not affect the number of nevi, but, to an extent, affected their average size, indicating that CD8+ T cells play some role in controlling the growth of nevi. CD8+ T cells are generated against melanoma antigens and are able to mount an efficient anti-melanoma response (24). Although others have proposed that IFNγ levels secreted by macrophages promote melanoma development, it is unclear whether low amounts of IFNγ are important to maintain an antitumor response or whether IFNγ is an antitumor agent during early stages of nevogenesis. In this study, we observed that in CD4KO mice there was an increase in IFNγ production and reduction in nevi numbers, indicating IFNγ may play an essential role in reducing early-oncogenic mutations, and, thus, nevus numbers. IFNγ is a pleiotropic molecule that is associated with antiproliferative, proapoptotic, and antitumor mechanisms. Although it is often considered as a major antitumor agent, and has been used in the treatment of many cancers due to its effective tumor immune surveillance. Type I IFNs play a role in DNA repair, and IFNγ early on treatment can induce cell-cycle arrest through Chk1 phosphorylation (25, 26).

However, many reports suggest that it may also play a protumorigenic role, because it can induce downregulation of MHC, and upregulation of indoleamine 2,3-dioxygenase and checkpoint inhibitors, such as programmed cell death ligand 1 (27, 28).

Previous studies suggest that the Langerhans cells present low doses of hapten, while dermal DCs present high doses of the hapten (29). Dermal CD103 DCs constitutively produce retinoic acid and have the potential to induce Foxp3+ Tregs (30). Our study did not investigate which antigen-presenting cell subset is responsible for inducing Tregs; however, we established that high DMBA doses induced expansion and the suppressive function of Tregs.

The mice treated with the larger amount of DMBA and that had increased numbers of nevi were also observed to have loss of p16 and p19 in a higher percentage of lesions than in those that were given smaller amounts. The CDKN2A locus encodes the two proteins, p16INK4a and p19ARF, the loss of which is critical for tumor development in many organ systems, including melanoma (31). Mutations in CDKN2A account for 35%–40% of familial melanomas (32). Melanocytic nevi persist in a benign state for years and seldom progress to melanoma even when mutations are present (33). High activation of H-RAS in Spitz nevi or mutated BRAF in melanocytes is associated with elevated p16INK4a expression (33, 34). Furthermore, it is well known that as nevi progress to melanomas, p16INK4a expression gradually decreases. Thus, there is a negative association between p16INK4a expression and melanoma progression (35).

The finding that deficiencies in immunologic function contribute to the development and growth of dysplastic nevi and potentially to their progression to melanoma is consistent with findings in humans. CD4+CD25+ T cells were increased 2-fold in metastatic LNs in comparison with both tumor-free LNs in patients with melanoma. These cells expressed the Foxp3 transcription factor and displayed an activated phenotype. These CD4+CD25+Foxp3+ cells were found to inhibit in vitro the proliferation and cytokine production of infiltrating effector CD4+CD25 and CD8+ T cells through a cell contact–dependent mechanism, thus behaving as Tregs (36). CD25+Foxp3+ Tregs were also prevalent both in junctional and compound atypical nevi and radial growth phase melanomas, suggesting that they can induce immunotolerance early during melanoma genesis, by positively regulating melanoma growth. The presence of these cells within a tumor site could be useful for prognosis and treatment of melanoma (37).

Organ transplant recipients on immunosuppressive therapy not only have a higher incidence of melanoma, but they also develop greater numbers of nevi as well (38–41). The nevi that develop in patients on immunosuppression have greater dermatoscopic changes than normal controls (41). Moreover, EMN frequently occur in the setting of immunosuppressive treatments (42). Treg therapies are in clinical trials to treat autoimmune diseases. If these results can be extrapolated to humans, then care should be taken when these immunosuppressive regimens are administered to individuals at increased risk for melanoma, and potentially other malignancies.

T.H. Nasti reports a patent for exosome-containing preparations from post-irradiated mouse melanoma cells and delay melanoma growth in vivo by a natural killer cell-dependent mechanism pending and a patent for benzodiazepine mediated radiosensitization of metastatic melanoma pending. No disclosures were reported by the other authors.

T.H. Nasti: Conceptualization, resources, supervision, funding acquisition, methodology, project administration, writing-review and editing. N. Yusuf: Conceptualization, resources, supervision, funding acquisition, methodology, project administration, writing-review and editing. M.A. Sherwani: Formal analysis, investigation, visualization, methodology. M. Athar: Conceptualization, resources, supervision, funding acquisition, project administration. L. Timares: Conceptualization, resources, data curation, formal analysis, supervision, validation, methodology, writing-original draft, writing-review and editing. C.A. Elmets: Conceptualization, resources, data curation, formal analysis, supervision, funding acquisition, validation, project administration, writing-review and editing.

This work was supported by NIH grant nos., P01CA210946, R01CA193885, P30 CA013148, N01 CN2012-00033, and 1R01AR071157- 01A1, and VA grant no., 101BX003395.

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.
Markovic
SN
,
Erickson
LA
,
Rao
RD
,
Weenig
RH
,
Pockaj
BA
,
Bardia
A
, et al
Malignant melanoma in the 21st century, part 2: staging, prognosis, and treatment
.
Mayo Clin Proc
2007
;
82
:
490
513
.
2.
Tucker
MA
,
Crutcher
WA
,
Hartge
P
,
Sagebiel
RW
. 
Familial and cutaneous features of dysplastic nevi: a case-control study
.
J Am Acad Dermatol
1993
;
28
:
558
64
.
3.
Alaibac
M
,
Piaserico
S
,
Rossi
CR
,
Foletto
M
,
Zacchello
G
,
Carli
P
, et al
Eruptive melanocytic nevi in patients with renal allografts: report of 10 cases with dermoscopic findings
.
J Am Acad Dermatol
2003
;
49
:
1020
2
.
4.
Gelfer
A
,
Rivers
JK
. 
Long-term follow-up of a patient with eruptive melanocytic nevi after Stevens-Johnson syndrome
.
Arch Dermatol
2007
;
143
:
1555
7
.
5.
John
JK
,
Smalley
KS
. 
Identification of BRAF mutations in eruptive melanocytic nevi: new insights into melanomagenesis?
Expert Rev Anticancer Ther
2011
;
11
:
711
4
.
6.
Hashimoto
M
,
Kamphorst
AO
,
Im
SJ
,
Kissick
HT
,
Pillai
RN
,
Ramalingam
SS
, et al
CD8 T cell exhaustion in chronic infection and cancer: opportunities for interventions
.
Annu Rev Med
2018
;
69
:
301
18
.
7.
Hodi
FS
,
O'Day
SJ
,
McDermott
DF
,
Weber
RW
,
Sosman
JA
,
Haanen
JB
, et al
Improved survival with ipilimumab in patients with metastatic melanoma
.
N Engl J Med
2010
;
363
:
711
23
.
8.
Robert
C
,
Long
GV
,
Brady
B
,
Dutriaux
C
,
Maio
M
,
Mortier
L
, et al
Nivolumab in previously untreated melanoma without BRAF mutation
.
N Engl J Med
2015
;
372
:
320
30
.
9.
Robert
C
,
Ribas
A
,
Wolchok
JD
,
Hodi
FS
,
Hamid
O
,
Kefford
R
, et al
Anti-programmed-death-receptor-1 treatment with pembrolizumab in ipilimumab-refractory advanced melanoma: a randomised dose-comparison cohort of a phase 1 trial
.
Lancet
2014
;
384
:
1109
17
.
10.
Nasti
TH
,
Cochran
JB
,
Tsuruta
Y
,
Yusuf
N
,
McKay
KM
,
Athar
M
, et al
A murine model for the development of melanocytic nevi and their progression to melanoma
.
Mol Carcinog
2016
;
55
:
646
58
.
11.
Yusuf
N
,
Nasti
TH
,
Katiyar
SK
,
Jacobs
MK
,
Seibert
MD
,
Ginsburg
AC
, et al
Antagonistic roles of CD4+ and CD8+ T-cells in 7,12-dimethylbenz(a)anthracene cutaneous carcinogenesis
.
Cancer Res
2008
;
68
:
3924
30
.
12.
Lecomte
C
,
Andujar
P
,
Renier
A
,
Kheuang
L
,
Abramowski
V
,
Mellottee
L
, et al
Similar tumor suppressor gene alteration profiles in asbestos‐induced murine and human mesothelioma
.
Cell Cycle
2005
;
4
:
1862
9
.
13.
Nasti
TH
,
Iqbal
O
,
Tamimi
IA
,
Geise
JT
,
Katiyar
SK
,
Yusuf
N
. 
Differential roles of T-cell subsets in regulation of ultraviolet radiation induced cutaneous photocarcinogenesis
.
Photochem Photobiol
2011
;
87
:
387
98
.
14.
Parsons
BL
,
McKinzie
PB
,
Heflich
RH
. 
Allele-specific competitive blocker-PCR detection of rare base substitution
.
Methods Mol Biol
2005
;
291
:
235
45
.
15.
Mehlman
MA
. 
Causal relationship from exposure to chemicals in oil refining and chemical industries and malignant melanoma
.
Ann N Y Acad Sci
2006
;
1076
:
822
8
.
16.
Schwarz
M
,
Munzel
PA
,
Braeuning
A
. 
Non-melanoma skin cancer in mouse and man
.
Arch Toxicol
2013
;
87
:
783
98
.
17.
Nasti
TH
,
Cochran
JB
,
Vachhani
RV
,
McKay
K
,
Tsuruta
Y
,
Athar
M
, et al
IL-23 inhibits melanoma development by augmenting DNA repair and modulating T cell subpopulations
.
J Immunol
2017
;
198
:
950
61
.
18.
Sakaguchi
S
,
Wing
K
,
Onishi
Y
,
Prieto-Martin
P
,
Yamaguchi
T
. 
Regulatory T cells: how do they suppress immune responses?
Int Immunol
2009
;
21
:
1105
11
.
19.
Baumgartner
J
,
Wilson
C
,
Palmer
B
,
Richter
D
,
Banerjee
A
,
McCarter
M
. 
Melanoma induces immunosuppression by up-regulating FOXP3(+) regulatory T cells
.
J Surg Res
2007
;
141
:
72
7
.
20.
Jang
JE
,
Hajdu
CH
,
Liot
C
,
Miller
G
,
Dustin
ML
,
Bar-Sagi
D
. 
Crosstalk between regulatory T cells and tumor-associated dendritic cells negates anti-tumor immunity in pancreatic cancer
.
Cell Rep
2017
;
20
:
558
71
.
21.
Jacobs
JF
,
Nierkens
S
,
Figdor
CG
,
de Vries
IJ
,
Adema
GJ
. 
Regulatory T cells in melanoma: the final hurdle towards effective immunotherapy?
Lancet Oncol
2012
;
13
:
e32
42
.
22.
Iversen
OH
. 
The skin tumorigenic and carcinogenic effects of different doses, numbers of dose fractions and concentrations of 7,12-dimethylbenz[a]anthracene in acetone applied on hairless mouse epidermis. Possible implications for human carcinogenesis
.
Carcinogenesis
1991
;
12
:
493
502
.
23.
Daniel
D
,
Meyer-Morse
N
,
Bergsland
EK
,
Dehne
K
,
Coussens
LM
,
Hanahan
D
. 
Immune enhancement of skin carcinogenesis by CD4+ T cells
.
J Exp Med
2003
;
197
:
1017
28
.
24.
Lozupone
F
,
Rivoltini
L
,
Luciani
F
,
Venditti
M
,
Lugini
L
,
Cova
A
, et al
Adoptive transfer of an anti-MART-1(27-35)-specific CD8+ T cell clone leads to immunoselection of human melanoma antigen-loss variants in SCID mice
.
Eur J Immunol
2003
;
33
:
556
66
.
25.
Brzostek-Racine
S
,
Gordon
C
,
Van Scoy
S
,
Reich
NC
. 
The DNA damage response induces IFN
.
J Immunol
2011
;
187
:
5336
45
.
26.
Kim
KS
,
Choi
KJ
,
Bae
S
. 
Interferon-gamma enhances radiation-induced cell death via downregulation of Chk1
.
Cancer Biol Ther
2012
;
13
:
1018
25
.
27.
Castro
F
,
Cardoso
AP
,
Goncalves
RM
,
Serre
K
,
Oliveira
MJ
. 
Interferon-gamma at the crossroads of tumor immune surveillance or evasion
.
Front Immunol
2018
;
9
:
847
.
28.
Cho
HI
,
Lee
YR
,
Celis
E
. 
Interferon γ limits the effectiveness of melanoma peptide vaccines
.
Blood
2011
;
117
:
135
44
.
29.
Bacci
S
,
Alard
P
,
Dai
R
,
Nakamura
T
,
Streilein
JW
. 
High and low doses of haptens dictate whether dermal or epidermal antigen-presenting cells promote contact hypersensitivity
.
Eur J Immunol
1997
;
27
:
442
8
.
30.
Guilliams
M
,
Crozat
K
,
Henri
S
,
Tamoutounour
S
,
Grenot
P
,
Devilard
E
, et al
Skin-draining lymph nodes contain dermis-derived CD103(-) dendritic cells that constitutively produce retinoic acid and induce Foxp3(+) regulatory T cells
.
Blood
2010
;
115
:
1958
68
.
31.
Quelle
DE
,
Cheng
M
,
Ashmun
RA
,
Sherr
CJ
. 
Cancer-associated mutations at the INK4a locus cancel cell cycle arrest by p16INK4a but not by the alternative reading frame protein p19ARF
.
Proc Natl Acad Sci U S A
1997
;
94
:
669
73
.
32.
Goldstein
AM
,
Chan
M
,
Harland
M
,
Gillanders
EM
,
Hayward
NK
,
Avril
MF
, et al
High-risk melanoma susceptibility genes and pancreatic cancer, neural system tumors, and uveal melanoma across GenoMEL
.
Cancer Res
2006
;
66
:
9818
28
.
33.
Michaloglou
C
,
Vredeveld
LC
,
Soengas
MS
,
Denoyelle
C
,
Kuilman
T
,
van der Horst
CM
, et al
BRAFE600-associated senescence-like cell cycle arrest of human naevi
.
Nature
2005
;
436
:
720
4
.
34.
Maldonado
JL
,
Timmerman
L
,
Fridlyand
J
,
Bastian
BC
. 
Mechanisms of cell-cycle arrest in Spitz nevi with constitutive activation of the MAP-kinase pathway
.
Am J Pathol
2004
;
164
:
1783
7
.
35.
Mihic-Probst
D
,
Mnich
CD
,
Oberholzer
PA
,
Seifert
B
,
Sasse
B
,
Moch
H
, et al
p16 expression in primary malignant melanoma is associated with prognosis and lymph node status
.
Int J Cancer
2006
;
118
:
2262
8
.
36.
Facciabene
A
,
Motz
GT
,
Coukos
G
. 
T-regulatory cells: key players in tumor immune escape and angiogenesis
.
Cancer Res
2012
;
72
:
2162
71
.
37.
Mourmouras
V
,
Fimiani
M
,
Rubegni
P
,
Epistolato
MC
,
Malagnino
V
,
Cardone
C
, et al
Evaluation of tumour-infiltrating CD4+CD25+FOXP3+ regulatory T cells in human cutaneous benign and atypical naevi, melanomas and melanoma metastases
.
Br J Dermatol
2007
;
157
:
531
9
.
38.
Bovenschen
HJ
,
Tjioe
M
,
Vermaat
H
,
de Hoop
D
,
Witteman
BM
,
Janssens
RW
, et al
Induction of eruptive benign melanocytic naevi by immune suppressive agents, including biologicals
.
Br J Dermatol
2006
;
154
:
880
4
.
39.
Smith
CH
,
McGregor
JM
,
Barker
JN
,
Morris
RW
,
Rigden
SP
,
MacDonald
DM
. 
Excess melanocytic nevi in children with renal allografts
.
J Am Acad Dermatol
1993
;
28
:
51
5
.
40.
Szepietowski
J
,
Wasik
F
,
Szepietowski
T
,
Włodarczyk
M
,
Sobczak-Radwan
K
,
Czyz
W
. 
Excess benign melanocytic naevi in renal transplant recipients
.
Dermatology
1997
;
194
:
17
9
.
41.
Koseoglu
G
,
Akay
BN
,
Kucuksahin
O
,
Erdem
C
. 
Dermoscopic changes in melanocytic nevi in patients receiving immunosuppressive and biologic treatments: results of a prospective case-control study
.
J Am Acad Dermatol
2015
;
73
:
623
9
.
42.
Vena
GA
,
Fargnoli
MC
,
Cassano
N
,
Argenziano
G
. 
Drug-induced eruptive melanocytic nevi
.
Expert Opin Drug Metab Toxicol
2017
;
13
:
293
300
.