Comparative immune phenotypic analysis of cutaneous Squamous Cell Carcinoma and Intraepidermal Carcinoma in immune-competent individuals: proportional representation of CD8+ T-cells but not FoxP3+ Regulatory T-cells is associated with disease stage

Andrew Freeman, Jennifer A Bridge, Pirashanthini Maruthayanar, Nana H Overgaard, Ji-Won Jung, Fiona Simpson, Tarl W Prow, H Peter Soyer, Ian H Frazer, Michael Freeman, James W Wells, Andrew Freeman, Jennifer A Bridge, Pirashanthini Maruthayanar, Nana H Overgaard, Ji-Won Jung, Fiona Simpson, Tarl W Prow, H Peter Soyer, Ian H Frazer, Michael Freeman, James W Wells

Abstract

Squamous Cell Carcinoma (SCC) is a type of non-melanoma skin cancer prevalent in immune-suppressed transplant recipients and older individuals with a history of chronic sun-exposure. SCC itself is believed to be a late-stage manifestation that can develop from premalignant lesions including Intraepidermal Carcinoma (IEC). Notably, while SCC regression is rare, IEC typically regresses in response to immune modifying topical treatments, however the underlying immunological reasons for these differential responses remain unclear. This study aimed to define whether IEC and SCC are associated with distinct immune profiles. We investigated the immune cell infiltrate of photo-damaged skin, IEC, and SCC tissue using 10-colour flow cytometry following fresh lesion digest. We found that IEC lesions contain higher percentages of CD3+ T-cells than photo-damaged skin, however, the abundance of CD3-CD56+ Natural Killer (NK) cells, CD11c+HLA-DR+ conventional Dendritic Cells (cDC), BDCA-2+HLA-DR+ plasmacytoid DC (pDC), FoxP3+ Regulatory T-cells (T-reg), Vα24+Vβ11+ invariant NKT-cells, and γδ Tcells did not alter with disease stage. Within the total T-cell population, high percentages of CD4+ T-cells were associated with SCC, yet CD8+ T-cells were less abundant in SCC compared with IEC. Our study demonstrates that while IEC lesions contain a higher proportion of T-cells than SCC lesions in general, SCC lesions specifically display a lower abundance of CD8+ T-cells than IEC. We propose that differences in CD8+ T-cell abundance contribute critically to the different capacity of SCC and IEC to regress in response to immune modifying topical treatments. Our study also suggests that a high ratio of CD4+ T-cells to CD8+ T-cells may be a immunological diagnostic indicator of late-stage SCC development in immune-competent patients.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Representative photomicrographs of each of…
Figure 1. Representative photomicrographs of each of the three analysis groups.
(A) Moderate to severe photo-damage in the upper dermis, (B) IEC, and (C) moderately differentiated SCC. Images presented are H&E stains, scale bar = 100 µm.
Figure 2. Gating strategy for analysis of…
Figure 2. Gating strategy for analysis of immune cell populations in tumour lesions by flow cytometry.
Age-matched sun-exposed skin or tumour lesions were excised and digested as outlined in Materials & Methods. Samples were stained with antibody and analysed by flow cytometry using the gating strategy shown in (A). (B) Ratio of Keratin 10+ Keratin 14− cells (suprabasal keratinocytes) to Keratin 10+ Keratin 14+ cells (basal keratinocytes) in the live CD45− population as determined by intracellular staining. (C) Comparison of viable immune cell extraction showing similar extraction efficacy between sample types. Live CD45+ cells as a percentage of the total live cell population is shown (skin; n = 6, IEC; n = 9, SCC; n = 13). ns: not significant. SSC-A: Side Scatter-Area, FSC-A: Forward Scatter-Area, TOF: Time of flight.
Figure 3. T-cells, NK cells, and DC…
Figure 3. T-cells, NK cells, and DC detected in photo-damaged skin and lesional skin of patients with IEC and SCC.
Fresh biopsies were digested to release cells and subsequently stained with antibodies to determine the representation of immune subpopulations by flow cytometry. (A) CD3+ T-cells (skin; n = 6, IEC; n = 9, SCC; n = 13), (B) CD3−CD56+ NK cells (skin; n = 6, IEC; n = 8, SCC; n = 13), (C) CD11c+HLA-DR+ cDC (includes both CD11bhi and CD11b−/lo populations, (skin; n = 6, IEC; n = 7, SCC; n = 7), (D) BDCA-2+HLA-DR+ pDC (skin; n = 6, IEC; n = 7, SCC; n = 7), (E) remaining CD11b+ myeloid cell populations (skin; n = 6, IEC; n = 7, SCC; n = 7), and (F) remaining immune cells (skin; n = 6, IEC; n = 6, SCC; n = 7). All data displayed are % of total CD45+ immune cell population. ns: not significant.
Figure 4. IEC and SCC lesions display…
Figure 4. IEC and SCC lesions display distinct patterns of CD4+ and CD8+ T-cell infiltration.
Fresh biopsies were digested to release cells and subsequently stained with antibodies to determine the representation of T-cell subpopulations by flow cytometry; (A) CD4+ T-cells (skin; n = 6, IEC; n = 9, SCC; n = 10), (B) FoxP3+ T-regs (skin; n = 6, IEC; n = 7, SCC; n = 8), (C) CD8+ T-cells (skin; n = 6, IEC; n = 8, SCC; n = 13), (D) TCRγδ+ T-cells (skin; n = 6, IEC; n = 9, SCC; n = 11), and (E) Vα24+Vβ11+ invariant Natural Killer T-cells (iNKT; (skin; n = 6, IEC; n = 9, SCC; n = 12). All data displayed are % of total CD3+ T-cells with the exception of (B) which represents the % of total CD3+CD4+ T-cells. ns: not significant.
Figure 5. SCC lesions display both an…
Figure 5. SCC lesions display both an increased CD4/CD8 ratio and an increased T-reg/CD8 ratio when compared with IEC lesions.
Absolute CD3+CD4+ T-cell, CD3+CD4+FoxP3+ T-cell, and CD3+CD8+ T-cell numbers within each lesion were determined by flow cytometry and used to determine the proportion of (A) CD3+CD4+ T-cells to CD3+CD8+ T-cells (skin; n = 6, IEC; n = 9, SCC; n = 11), and (B) CD3+CD4+FoxP3+ T-cells to CD3+CD8+ T-cells (skin; n = 6, IEC; n = 7, SCC; n = 9). ns: not significant.

References

    1. Ziegler A, Jonason AS, Leffell DJ, Simon JA, Sharma HW, et al. (1994) Sunburn and p53 in the onset of skin cancer. Nature 372: 773–776.
    1. Anwar J, Wrone DA, Kimyai-Asadi A, Alam M (2004) The development of actinic keratosis into invasive squamous cell carcinoma: evidence and evolving classification schemes. Clinics in dermatology 22: 189–196.
    1. Dodson JM, DeSpain J, Hewett JE, Clark DP (1991) Malignant potential of actinic keratoses and the controversy over treatment. A patient-oriented perspective. Archives of dermatology 127: 1029–1031.
    1. Marks R, Foley P, Goodman G, Hage BH, Selwood TS (1986) Spontaneous remission of solar keratoses: the case for conservative management. The British journal of dermatology 115: 649–655.
    1. Ulrich C, Schmook T, Nindl I, Meyer T, Sterry W, et al. (2003) Cutaneous precancers in organ transplant recipients: an old enemy in a new surrounding. The British journal of dermatology 149 Suppl 6640–42.
    1. Vajdic CM, van Leeuwen MT (2009) Cancer incidence and risk factors after solid organ transplantation. International journal of cancer Journal international du cancer 125: 1747–1754.
    1. Akkilic-Materna M, Massone C, Komericki P (2011) Imiquimod and lymphatic field clearance: a new hypothesis based on a remote immune action on skin cancer. Acta dermato-venereologica 91: 432–435.
    1. Hanke CW, Swanson N, Bruce S, Berman B, Kulp J, et al. (2011) Complete clearance is sustained for at least 12 months after treatment of actinic keratoses of the face or balding scalp via daily dosing with imiquimod 3.75% or 2.5% cream. Journal of drugs in dermatology: JDD 10: 165–170.
    1. Ulrich C, Johannsen A, Rowert-Huber J, Ulrich M, Sterry W, et al. (2010) Results of a randomized, placebo-controlled safety and efficacy study of topical diclofenac 3% gel in organ transplant patients with multiple actinic keratoses. European journal of dermatology: EJD 20: 482–488.
    1. Love WE, Bernhard JD, Bordeaux JS (2009) Topical imiquimod or fluorouracil therapy for basal and squamous cell carcinoma: a systematic review. Archives of dermatology 145: 1431–1438.
    1. Beham A, Regauer S, Soyer HP, Beham-Schmid C (1998) Keratoacanthoma: a clinically distinct variant of well differentiated squamous cell carcinoma. Adv Anat Pathol 5: 269–280.
    1. Harnden P, Southgate J (1997) Cytokeratin 14 as a marker of squamous differentiation in transitional cell carcinomas. J Clin Pathol 50: 1032–1033.
    1. Chu PG, Lyda MH, Weiss LM (2001) Cytokeratin 14 expression in epithelial neoplasms: a survey of 435 cases with emphasis on its value in differentiating squamous cell carcinomas from other epithelial tumours. Histopathology 39: 9–16.
    1. Hussein MR, Ahmed RA (2005) Analysis of the mononuclear inflammatory cell infiltrate in the non-tumorigenic, pre-tumorigenic and tumorigenic keratinocytic hyperproliferative lesions of the skin. Cancer biology & therapy 4: 819–821.
    1. Clark RA, Chong BF, Mirchandani N, Yamanaka K, Murphy GF, et al. (2006) A novel method for the isolation of skin resident T cells from normal and diseased human skin. The Journal of investigative dermatology 126: 1059–1070.
    1. Zhang S, Fujita H, Mitsui H, Yanofsky VR, Fuentes-Duculan J, et al. (2013) Increased Tc22 and Treg/CD8 ratio contribute to aggressive growth of transplant associated squamous cell carcinoma. PLoS One 8: e62154.
    1. Junankar SR, Eichten A, Kramer A, de Visser KE, Coussens LM (2006) Analysis of immune cell infiltrates during squamous carcinoma development. J Investig Dermatol Symp Proc 11: 36–43.
    1. Clark RA, Huang SJ, Murphy GF, Mollet IG, Hijnen D, et al. (2008) Human squamous cell carcinomas evade the immune response by down-regulation of vascular E-selectin and recruitment of regulatory T cells. The Journal of experimental medicine 205: 2221–2234.
    1. Kosmidis M, Dziunycz P, Suarez-Farinas M, Muhleisen B, Scharer L, et al. (2010) Immunosuppression affects CD4+ mRNA expression and induces Th2 dominance in the microenvironment of cutaneous squamous cell carcinoma in organ transplant recipients. J Immunother 33: 538–546.
    1. Jang TJ (2008) Prevalence of Foxp3 positive T regulatory cells is increased during progression of cutaneous squamous tumors. Yonsei medical journal 49: 942–948.
    1. Berhane T, Halliday GM, Cooke B, Barnetson RS (2002) Inflammation is associated with progression of actinic keratoses to squamous cell carcinomas in humans. Br J Dermatol 146: 810–815.
    1. Smolle J, Soyer HP, Ehall R, Stettner H, Kerl H (1987) NK cell density in malignant skin tumours–a stereological study. Br J Dermatol 116: 823–829.
    1. Bier H, Hoffmann T, Haas I, van Lierop A (1998) Anti-(epidermal growth factor) receptor monoclonal antibodies for the induction of antibody-dependent cell-mediated cytotoxicity against squamous cell carcinoma lines of the head and neck. Cancer Immunol Immunother 46: 167–173.
    1. Muhleisen B, Petrov I, Gachter T, Kurrer M, Scharer L, et al. (2009) Progression of cutaneous squamous cell carcinoma in immunosuppressed patients is associated with reduced CD123+ and FOXP3+ cells in the perineoplastic inflammatory infiltrate. Histopathology 55: 67–76.
    1. Halliday GM, Patel A, Hunt MJ, Tefany FJ, Barnetson RS (1995) Spontaneous regression of human melanoma/nonmelanoma skin cancer: association with infiltrating CD4+ T cells. World journal of surgery 19: 352–358.
    1. Patel A, Halliday GM, Barnetson RS (1995) CD4+ T lymphocyte infiltration correlates with regression of a UV-induced squamous cell carcinoma. J Dermatol Sci 9: 12–19.
    1. Peguillet I, Milder M, Louis D, Vincent-Salomon A, Dorval T, et al. (2014) High numbers of differentiated effector CD4 T cells are found in patients with cancer and correlate with clinical response after neoadjuvant therapy of breast cancer. Cancer Res 74: 2204–2216.
    1. Bluth MJ, Zaba LC, Moussai D, Suarez-Farinas M, Kaporis H, et al. (2009) Myeloid dendritic cells from human cutaneous squamous cell carcinoma are poor stimulators of T-cell proliferation. J Invest Dermatol 129: 2451–2462.
    1. Carroll RP, Segundo DS, Hollowood K, Marafioti T, Clark TG, et al. (2010) Immune phenotype predicts risk for posttransplantation squamous cell carcinoma. J Am Soc Nephrol 21: 713–722.
    1. Hope CM, Grace BS, Pilkington KR, Coates PT, Bergmann IP, et al... (2014) The immune phenotype may relate to cancer development in kidney transplant recipients. Kidney Int.
    1. Fukunaga A, Khaskhely NM, Ma Y, Sreevidya CS, Taguchi K, et al. (2010) Langerhans cells serve as immunoregulatory cells by activating NKT cells. Journal of immunology 185: 4633–4640.
    1. Moodycliffe AM, Nghiem D, Clydesdale G, Ullrich SE (2000) Immune suppression and skin cancer development: regulation by NKT cells. Nature immunology 1: 521–525.
    1. Mattarollo SR, Rahimpour A, Choyce A, Godfrey DI, Leggatt GR, et al. (2010) Invariant NKT cells in hyperplastic skin induce a local immune suppressive environment by IFN-gamma production. Journal of immunology 184: 1242–1250.
    1. Mattarollo SR, Yong M, Gosmann C, Choyce A, Chan D, et al. (2011) NKT cells inhibit antigen-specific effector CD8 T cell induction to skin viral proteins. Journal of immunology 187: 1601–1608.
    1. Watarai H, Nakagawa R, Omori-Miyake M, Dashtsoodol N, Taniguchi M (2008) Methods for detection, isolation and culture of mouse and human invariant NKT cells. Nature protocols 3: 70–78.
    1. Sarchio SN, Scolyer RA, Beaugie C, McDonald D, Marsh-Wakefield F, et al. (2014) Pharmacologically antagonizing the CXCR4-CXCL12 chemokine pathway with AMD3100 inhibits sunlight-induced skin cancer. J Invest Dermatol 134: 1091–1100.
    1. Kirshenbaum AS, Goff JP, Semere T, Foster B, Scott LM, et al. (1999) Demonstration that human mast cells arise from a progenitor cell population that is CD34(+), c-kit(+), and expresses aminopeptidase N (CD13). Blood 94: 2333–2342.
    1. Parizi AC, Barbosa RL, Parizi JL, Nai GA (2010) A comparison between the concentration of mast cells in squamous cell carcinomas of the skin and oral cavity. An Bras Dermatol 85: 811–818.

Source: PubMed

3
Se inscrever